1
|
Shih MFM, Zhang J, Brown EB, Dubnau J, Keene AC. Targeted single cell expression profiling identifies integrators of sleep and metabolic state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614841. [PMID: 39386468 PMCID: PMC11463630 DOI: 10.1101/2024.09.25.614841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Animals modulate sleep in accordance with their internal and external environments. Metabolic cues are particularly potent regulators of sleep, allowing animals to alter their sleep timing and amount depending on food availability and foraging duration. The fruit fly, Drosophila melanogaster, suppresses sleep in response to acute food deprivation, presumably to forage for food. This process is dependent on a single pair of Lateral Horn Leucokinin (LHLK) neurons, that secrete the neuropeptide Leucokinin. These neurons signal to insulin producing cells and suppress sleep under periods of starvation. The identification of individual neurons that modulate sleep-metabolism interactions provides the opportunity to examine the cellular changes associated with sleep modulation. Here, we use single-cell sequencing of LHLK neurons to examine the transcriptional responses to starvation. We validate that a Patch-seq approach selectively isolates RNA from individual LHLK neurons. Single-cell CEL-Seq comparisons of LHLK neurons between fed and 24-hr starved flies identified 24 genes that are differentially expressed in accordance with starvation state. In total, 12 upregulated genes and 12 downregulated genes were identified. Gene-ontology analysis showed an enrichment for Attacins, a family of anti-microbial peptides, along with several transcripts with diverse roles in regulating cellular function. Targeted knockdown of differentially expressed genes identified multiple genes that function within LHLK neurons to regulate sleep-metabolism interactions. Functionally validated genes include an essential role for the E3 ubiquitin Ligase insomniac, the sorbitol dehydrogenase Sodh1, as well as AttacinC and AttacinB in starvation-induced sleep suppression. Taken together, these findings provide a pipeline for identifying novel regulators of sleep-metabolism interactions within individual neurons.
Collapse
Affiliation(s)
| | - Jiwei Zhang
- Department of Biology, Texas A&M University, College Station, TX 77840
| | | | - Joshua Dubnau
- Dept of Anesthesiology, Stony Brook School of Medicine, Stony Brook NY, 11794
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook NY, 11794
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, TX 77840
| |
Collapse
|
2
|
Lv P, Zhao Z, Hirano Y, Du J. The CoREST complex regulates multiple histone modifications temporal-specifically in clock neurons. Open Biol 2024; 14:230355. [PMID: 38981515 DOI: 10.1098/rsob.230355] [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: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 07/11/2024] Open
Abstract
Epigenetic regulation is important for circadian rhythm. In previous studies, multiple histone modifications were found at the Period (Per) locus. However, most of these studies were not conducted in clock neurons. In our screen, we found that a CoREST mutation resulted in defects in circadian rhythm by affecting Per transcription. Based on previous studies, we hypothesized that CoREST regulates circadian rhythm by regulating multiple histone modifiers at the Per locus. Genetic and physical interaction experiments supported these regulatory relationships. Moreover, through tissue-specific chromatin immunoprecipitation assays in clock neurons, we found that the CoREST mutation led to time-dependent changes in corresponding histone modifications at the Per locus. Finally, we proposed a model indicating the role of the CoREST complex in the regulation of circadian rhythm. This study revealed the dynamic changes of histone modifications at the Per locus specifically in clock neurons. Importantly, it provides insights into the role of epigenetic factors in the regulation of dynamic gene expression changes in circadian rhythm.
Collapse
Affiliation(s)
- Pengfei Lv
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, People's Republic of China
| | - Zhangwu Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, People's Republic of China
| | - Yukinori Hirano
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Juan Du
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, People's Republic of China
| |
Collapse
|
3
|
Zhao X, Yang X, Lv P, Xu Y, Wang X, Zhao Z, Du J. Polycomb regulates circadian rhythms in Drosophila in clock neurons. Life Sci Alliance 2024; 7:e202302140. [PMID: 37914396 PMCID: PMC10620068 DOI: 10.26508/lsa.202302140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023] Open
Abstract
Circadian rhythms are essential physiological feature for most living organisms. Previous studies have shown that epigenetic regulation plays a crucial role. There is a knowledge gap in the chromatin state of some key clock neuron clusters. In this study, we show that circadian rhythm is affected by the epigenetic regulator Polycomb (Pc) within the Drosophila clock neurons. To investigate the molecular mechanisms underlying the roles of Pc in these clock neuron clusters, we use targeted DamID (TaDa) to identify genes significantly bound by Pc in the neurons marked by C929-Gal4 (including l-LNvs cluster), R6-Gal4 (including s-LNvs cluster), R18H11-Gal4 (including DN1 cluster), and DVpdf-Gal4, pdf-Gal80 (including LNds cluster). It shows that Pc binds to the genes involved in the circadian rhythm pathways, arguing a direct role for Pc in regulating circadian rhythms through specific clock genes. This study shows the identification of Pc targets in the clock neuron clusters, providing potential resource for understanding the regulatory mechanisms of circadian rhythms by the PcG complex. Thus, this study provided an example for epigenetic regulation of adult behavior.
Collapse
Affiliation(s)
- Xianguo Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xingzhuo Yang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Pengfei Lv
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yuetong Xu
- Department of Crop Genomics and Bioinformatics, College of Agronomy and Biotechnology, National Maize Improvement Center of China, China Agricultural University, Beijing, China
| | - Xiangfeng Wang
- Department of Crop Genomics and Bioinformatics, College of Agronomy and Biotechnology, National Maize Improvement Center of China, China Agricultural University, Beijing, China
| | - Zhangwu Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Juan Du
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
4
|
Jovanoski KD, Duquenoy L, Mitchell J, Kapoor I, Treiber CD, Croset V, Dempsey G, Parepalli S, Cognigni P, Otto N, Felsenberg J, Waddell S. Dopaminergic systems create reward seeking despite adverse consequences. Nature 2023; 623:356-365. [PMID: 37880370 PMCID: PMC10632144 DOI: 10.1038/s41586-023-06671-8] [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: 07/04/2022] [Accepted: 09/22/2023] [Indexed: 10/27/2023]
Abstract
Resource-seeking behaviours are ordinarily constrained by physiological needs and threats of danger, and the loss of these controls is associated with pathological reward seeking1. Although dysfunction of the dopaminergic valuation system of the brain is known to contribute towards unconstrained reward seeking2,3, the underlying reasons for this behaviour are unclear. Here we describe dopaminergic neural mechanisms that produce reward seeking despite adverse consequences in Drosophila melanogaster. Odours paired with optogenetic activation of a defined subset of reward-encoding dopaminergic neurons become cues that starved flies seek while neglecting food and enduring electric shock punishment. Unconstrained seeking of reward is not observed after learning with sugar or synthetic engagement of other dopaminergic neuron populations. Antagonism between reward-encoding and punishment-encoding dopaminergic neurons accounts for the perseverance of reward seeking despite punishment, whereas synthetic engagement of the reward-encoding dopaminergic neurons also impairs the ordinary need-dependent dopaminergic valuation of available food. Connectome analyses reveal that the population of reward-encoding dopaminergic neurons receives highly heterogeneous input, consistent with parallel representation of diverse rewards, and recordings demonstrate state-specific gating and satiety-related signals. We propose that a similar dopaminergic valuation system dysfunction is likely to contribute to maladaptive seeking of rewards by mammals.
Collapse
Affiliation(s)
| | - Lucille Duquenoy
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Jessica Mitchell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ishaan Kapoor
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | | | - Vincent Croset
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Department of Biosciences, Durham University, Durham, UK
| | - Georgia Dempsey
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Sai Parepalli
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Paola Cognigni
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Northern Medical Physics and Clinical Engineering, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Nils Otto
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster, Germany
| | - Johannes Felsenberg
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Genome-wide analysis identifies Homothorax and Extradenticle as regulators of insulin in Drosophila Insulin-Producing cells. PLoS Genet 2022; 18:e1010380. [PMID: 36095003 PMCID: PMC9499297 DOI: 10.1371/journal.pgen.1010380] [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: 01/27/2022] [Revised: 09/22/2022] [Accepted: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Drosophila Insulin-Producing Cells (IPCs) are the main production site of the Drosophila Insulin-like peptides or dilps which have key roles in regulating growth, development, reproduction, lifespan and metabolism. To better understand the signalling pathways and transcriptional networks that are active in the IPCs we queried publicly available transcriptome data of over 180 highly inbred fly lines for dilp expression and used dilp expression as the input for a Genome-wide association study (GWAS). This resulted in the identification of variants in 125 genes that were associated with variation in dilp expression. The function of 57 of these genes in the IPCs was tested using an RNAi-based approach. We found that IPC-specific depletion of most genes resulted in differences in expression of one or more of the dilps. We then elaborated further on one of the candidate genes with the strongest effect on dilp expression, Homothorax, a transcription factor known for its role in eye development. We found that Homothorax and its binding partner Extradenticle are involved in regulating dilp2, -3 and -5 expression and that genetic depletion of both TFs shows phenotypes associated with reduced insulin signalling. Furthermore, we provide evidence that other transcription factors involved in eye development are also functional in the IPCs. In conclusion, we showed that this expression level-based GWAS approach identified genetic regulators implicated in IPC function and dilp expression. Insulin signalling has a central and evolutionarily conserved role in many processes including growth, development, reproduction, lifespan, stress resistance and metabolic homeostasis. In the fruitfly Drosophila melanogaster insulin-producing cells in the brain are the main source of three insulin-like peptides, Dilp2, -3 and -5. How the production and secretion of these three insulin-like peptides are regulated remains incompletely understood. In the current study, genome-wide association studies were used to identify 50 novel regulators of Dilp2, -3 and -5. We show that one of the top candidate regulators, Homothorax, is an important regulator of dilp2, -3 and –5 expression in the IPCs and is necessary for normal systemic insulin signalling and regulates adult size and developmental timing. We also show that the Hth interactor Extradenticle (Exd) is equally required in the adult but not in the larval IPCs. Finally, we show that most genes of the so-called retinal determination gene network are expressed in the IPCs and regulate normal dilp2 and -5 expression. Together, these results identify further regulatory levels active in the IPCs and implicate a reshuffled version of a previously identified gene regulatory network therein.
Collapse
|
6
|
Crespo-Flores SL, Barber AF. The Drosophila circadian clock circuit is a nonhierarchical network of peptidergic oscillators. CURRENT OPINION IN INSECT SCIENCE 2022; 52:100944. [PMID: 35709899 DOI: 10.1016/j.cois.2022.100944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
The relatively simple Drosophila circadian clock circuit consists of 150 clock neurons that coordinate rhythmic behavior and physiology, which are generally classified based on neuroanatomical location. Transcriptional and connectomic studies have identified novel subdivisions of these clock neuron populations, and identified neuropeptides not previously known to be expressed in the fly clock circuit. An additional feature of fly clock neurons is daily axonal remodeling, first noted in small ventrolateral neurons, but more recently also found in additional clock neuron groups. These findings raise new questions about the functional roles of clock neuron subpopulations and daily remodeling of network architecture in regulating circadian behavior and physiology.
Collapse
Affiliation(s)
- Sergio L Crespo-Flores
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers, the State University of New Jersey, USA
| | - Annika F Barber
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers, the State University of New Jersey, USA.
| |
Collapse
|
7
|
Damage-responsive neuro-glial clusters coordinate the recruitment of dormant neural stem cells in Drosophila. Dev Cell 2022; 57:1661-1675.e7. [PMID: 35716661 DOI: 10.1016/j.devcel.2022.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022]
Abstract
Recruitment of stem cells is crucial for tissue repair. Although stem cell niches can provide important signals, little is known about mechanisms that coordinate the engagement of disseminated stem cells across an injured tissue. In Drosophila, adult brain lesions trigger local recruitment of scattered dormant neural stem cells suggesting a mechanism for creating a transient stem cell activation zone. Here, we find that injury triggers a coordinated response in neuro-glial clusters that promotes the spread of a neuron-derived stem cell factor via glial secretion of the lipocalin-like transporter Swim. Strikingly, swim is induced in a Hif1-α-dependent manner in response to brain hypoxia. Mammalian Swim (Lcn7) is also upregulated in glia of the mouse hippocampus upon brain injury. Our results identify a central role of neuro-glial clusters in promoting neural stem cell activation at a distance, suggesting a conserved function of the HIF1-α/Swim/Wnt module in connecting injury-sensing and regenerative outcomes.
Collapse
|
8
|
Reinhard N, Schubert FK, Bertolini E, Hagedorn N, Manoli G, Sekiguchi M, Yoshii T, Rieger D, Helfrich-Förster C. The Neuronal Circuit of the Dorsal Circadian Clock Neurons in Drosophila melanogaster. Front Physiol 2022; 13:886432. [PMID: 35574472 PMCID: PMC9100938 DOI: 10.3389/fphys.2022.886432] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022] Open
Abstract
Drosophila’s dorsal clock neurons (DNs) consist of four clusters (DN1as, DN1ps, DN2s, and DN3s) that largely differ in size. While the DN1as and the DN2s encompass only two neurons, the DN1ps consist of ∼15 neurons, and the DN3s comprise ∼40 neurons per brain hemisphere. In comparison to the well-characterized lateral clock neurons (LNs), the neuroanatomy and function of the DNs are still not clear. Over the past decade, numerous studies have addressed their role in the fly’s circadian system, leading to several sometimes divergent results. Nonetheless, these studies agreed that the DNs are important to fine-tune activity under light and temperature cycles and play essential roles in linking the output from the LNs to downstream neurons that control sleep and metabolism. Here, we used the Flybow system, specific split-GAL4 lines, trans-Tango, and the recently published fly connectome (called hemibrain) to describe the morphology of the DNs in greater detail, including their synaptic connections to other clock and non-clock neurons. We show that some DN groups are largely heterogenous. While certain DNs are strongly connected with the LNs, others are mainly output neurons that signal to circuits downstream of the clock. Among the latter are mushroom body neurons, central complex neurons, tubercle bulb neurons, neurosecretory cells in the pars intercerebralis, and other still unidentified partners. This heterogeneity of the DNs may explain some of the conflicting results previously found about their functionality. Most importantly, we identify two putative novel communication centers of the clock network: one fiber bundle in the superior lateral protocerebrum running toward the anterior optic tubercle and one fiber hub in the posterior lateral protocerebrum. Both are invaded by several DNs and LNs and might play an instrumental role in the clock network.
Collapse
Affiliation(s)
- Nils Reinhard
- Julius Maximilian University of Würzburg, Würzburg, Germany
| | | | - Enrico Bertolini
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Würzburg, Germany
| | | | - Giulia Manoli
- Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Manabu Sekiguchi
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Taishi Yoshii
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Dirk Rieger
- Julius Maximilian University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
9
|
Miozzo F, Valencia-Alarcón EP, Stickley L, Majcin Dorcikova M, Petrelli F, Tas D, Loncle N, Nikonenko I, Bou Dib P, Nagoshi E. Maintenance of mitochondrial integrity in midbrain dopaminergic neurons governed by a conserved developmental transcription factor. Nat Commun 2022; 13:1426. [PMID: 35301315 PMCID: PMC8931002 DOI: 10.1038/s41467-022-29075-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/25/2022] [Indexed: 12/21/2022] Open
Abstract
Progressive degeneration of dopaminergic (DA) neurons in the substantia nigra is a hallmark of Parkinson’s disease (PD). Dysregulation of developmental transcription factors is implicated in dopaminergic neurodegeneration, but the underlying molecular mechanisms remain largely unknown. Drosophila Fer2 is a prime example of a developmental transcription factor required for the birth and maintenance of midbrain DA neurons. Using an approach combining ChIP-seq, RNA-seq, and genetic epistasis experiments with PD-linked genes, here we demonstrate that Fer2 controls a transcriptional network to maintain mitochondrial structure and function, and thus confers dopaminergic neuroprotection against genetic and oxidative insults. We further show that conditional ablation of Nato3, a mouse homolog of Fer2, in differentiated DA neurons causes mitochondrial abnormalities and locomotor impairments in aged mice. Our results reveal the essential and conserved role of Fer2 homologs in the mitochondrial maintenance of midbrain DA neurons, opening new perspectives for modeling and treating PD. Mitochondrial dysfunction in dopaminergic neurons is a pathological hallmark of Parkinson’s disease. Here, the authors find a conserved mechanism by which a single transcription factor controls mitochondrial health in dopaminergic neurons during the aging process.
Collapse
Affiliation(s)
- Federico Miozzo
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland.,Neuroscience Institute - CNR (IN-CNR), Milan, Italy
| | - Eva P Valencia-Alarcón
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland
| | - Luca Stickley
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland
| | - Michaëla Majcin Dorcikova
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland
| | | | - Damla Tas
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland.,The Janssen Pharmaceutical Companies of Johnson & Johnson, Bern, Switzerland
| | - Nicolas Loncle
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland.,Puma Biotechnology, Inc., Berkeley, CA, USA
| | - Irina Nikonenko
- Department of Basic Neurosciences and the Center for Neuroscience, CMU, University of Geneva, CH-1211, Geneva 4, Switzerland
| | - Peter Bou Dib
- Institute of Cell Biology, University of Bern, CH-3012, Bern, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1211, Geneva 4, Switzerland.
| |
Collapse
|
10
|
Sato K, Yamamoto D. Mutually exclusive expression of sex-specific and non-sex-specific fruitless gene products in the Drosophila central nervous system. Gene Expr Patterns 2022; 43:119232. [DOI: 10.1016/j.gep.2022.119232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/19/2022] [Accepted: 01/28/2022] [Indexed: 11/04/2022]
|
11
|
Yang N, Srivastav SP, Rahman R, Ma Q, Dayama G, Li S, Chinen M, Lei EP, Rosbash M, Lau NC. Transposable element landscapes in aging Drosophila. PLoS Genet 2022; 18:e1010024. [PMID: 35239675 PMCID: PMC8893327 DOI: 10.1371/journal.pgen.1010024] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 11/28/2022] Open
Abstract
Genetic mechanisms that repress transposable elements (TEs) in young animals decline during aging, as reflected by increased TE expression in aged animals. Does increased TE expression during aging lead to more genomic TE copies in older animals? To address this question, we quantified TE Landscapes (TLs) via whole genome sequencing of young and aged Drosophila strains of wild-type and mutant backgrounds. We quantified TLs in whole flies and dissected brains and validated the feasibility of our approach in detecting new TE insertions in aging Drosophila genomes when small RNA and RNA interference (RNAi) pathways are compromised. We also describe improved sequencing methods to quantify extra-chromosomal DNA circles (eccDNAs) in Drosophila as an additional source of TE copies that accumulate during aging. Lastly, to combat the natural progression of aging-associated TE expression, we show that knocking down PAF1, a conserved transcription elongation factor that antagonizes RNAi pathways, may bolster suppression of TEs during aging and extend lifespan. Our study suggests that in addition to a possible influence by different genetic backgrounds, small RNA and RNAi mechanisms may mitigate genomic TL expansion despite the increase in TE transcripts during aging. Transposable elements, also called transposons, are genetic parasites found in all animal genomes. Normally, transposons are compacted away in silent chromatin in young animals. But, as animals age and transposon-silencing defense mechanisms break down, transposon RNAs accumulate to significant levels in old animals like fruit flies. An open question is whether the increased levels of transposon RNAs in older animals also correspond to increased genomic copies of transposons. This study approached this question by sequencing the whole genomes of young and old wild-type and mutant flies lacking a functional RNA interference (RNAi) pathway, which naturally silences transposon RNAs. Although the wild-type flies with intact RNAi activity had little new accumulation of transposon copies, the sequencing approach was able to detect several transposon accumulation occurrences in some RNAi mutants. In addition, we found that some fly transposon families can also accumulate as extra-chromosomal circular DNA copies. Lastly, we showed that genetically augmenting the expression of RNAi factors can counteract the rising transposon RNA levels in aging and promote longevity. This study improves our understanding of the animal host genome relationship with transposons during natural aging processes.
Collapse
Affiliation(s)
- Nachen Yang
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
| | - Satyam P. Srivastav
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
| | - Reazur Rahman
- Brandeis University, Department of Biology and Howard Hughes Medical Institute, Waltham, Massachusetts, United States of America
| | - Qicheng Ma
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
| | - Gargi Dayama
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
| | - Sizheng Li
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
| | - Madoka Chinen
- Nuclear Organization and Gene Expression Section, NIDDK, NIH, Bethesda, Maryland, United States of America
| | - Elissa P. Lei
- Nuclear Organization and Gene Expression Section, NIDDK, NIH, Bethesda, Maryland, United States of America
| | - Michael Rosbash
- Brandeis University, Department of Biology and Howard Hughes Medical Institute, Waltham, Massachusetts, United States of America
| | - Nelson C. Lau
- Boston University School of Medicine, Department of Biochemistry, Boston, Massachusetts, United States of America
- Boston University Genome Science Institute, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
12
|
Systematic modeling-driven experiments identify distinct molecular clockworks underlying hierarchically organized pacemaker neurons. Proc Natl Acad Sci U S A 2022; 119:2113403119. [PMID: 35193959 PMCID: PMC8872709 DOI: 10.1073/pnas.2113403119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Abstract
In metazoan organisms, circadian (∼24 h) rhythms are regulated by pacemaker neurons organized in a master-slave hierarchy. Although it is widely accepted that master pacemakers and slave oscillators generate rhythms via an identical negative feedback loop of transcription factor CLOCK (CLK) and repressor PERIOD (PER), their different roles imply heterogeneity in their molecular clockworks. Indeed, in Drosophila, defective binding between CLK and PER disrupts molecular rhythms in the master pacemakers, small ventral lateral neurons (sLNvs), but not in the slave oscillator, posterior dorsal neuron 1s (DN1ps). Here, we develop a systematic and expandable approach that unbiasedly searches the source of the heterogeneity in molecular clockworks from time-series data. In combination with in vivo experiments, we find that sLNvs exhibit higher synthesis and turnover of PER and lower CLK levels than DN1ps. Importantly, light shift analysis reveals that due to such a distinct molecular clockwork, sLNvs can obtain paradoxical characteristics as the master pacemaker, generating strong rhythms that are also flexibly adjustable to environmental changes. Our results identify the different characteristics of molecular clockworks of pacemaker neurons that underlie hierarchical multi-oscillator structure to ensure the rhythmic fitness of the organism.
Collapse
|
13
|
van Alphen B, Stewart S, Iwanaszko M, Xu F, Li K, Rozenfeld S, Ramakrishnan A, Itoh TQ, Sisobhan S, Qin Z, Lear BC, Allada R. Glial immune-related pathways mediate effects of closed head traumatic brain injury on behavior and lethality in Drosophila. PLoS Biol 2022; 20:e3001456. [PMID: 35081110 PMCID: PMC8791498 DOI: 10.1371/journal.pbio.3001456] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.
Collapse
Affiliation(s)
- Bart van Alphen
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Samuel Stewart
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Marta Iwanaszko
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- Department of Preventive Medicine—Biostatistics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Keyin Li
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Sydney Rozenfeld
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Anujaianthi Ramakrishnan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Taichi Q. Itoh
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Shiju Sisobhan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Zuoheng Qin
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Bridget C. Lear
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
14
|
Shegelski VA, Evenden ML, Huber DPW, Sperling FAH. Identification of genes and gene expression associated with dispersal capacity in the mountain pine beetle, Dendroctonus ponderosae Hopkins (Coleoptera: Curculionidae). PeerJ 2021; 9:e12382. [PMID: 34754626 PMCID: PMC8555496 DOI: 10.7717/peerj.12382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022] Open
Abstract
Dispersal flights by the mountain pine beetle have allowed range expansion and major damage to pine stands in western Canada. We asked what the genetic and transcriptional basis of mountain pine beetle dispersal capacity is. Using flight mills, RNA-seq and a targeted association study, we compared strong-flying, weak-flying, and non-flying female beetles from the recently colonized northern end of their range. Nearly 3,000 genes were differentially expressed between strong and weak flying beetles, while weak fliers and nonfliers did not significantly differ. The differentially expressed genes were mainly associated with lipid metabolism, muscle maintenance, oxidative stress response, detoxification, endocrine function, and flight behavior. Three variant loci, two in the coding region of genes, were significantly associated with flight capacity but these genes had no known functional link to flight. Several differentially expressed gene systems may be important for sustained flight, while other systems are downregulated during dispersal and likely to conserve energy before host colonization. The candidate genes and SNPs identified here will inform further studies and management of mountain pine beetle, as well as contribute to understanding the mechanisms of insect dispersal flights.
Collapse
Affiliation(s)
- Victor A Shegelski
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Maya L Evenden
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Dezene P W Huber
- Faculty of Environment, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Felix A H Sperling
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Machado Almeida P, Lago Solis B, Stickley L, Feidler A, Nagoshi E. Neurofibromin 1 in mushroom body neurons mediates circadian wake drive through activating cAMP-PKA signaling. Nat Commun 2021; 12:5758. [PMID: 34599173 PMCID: PMC8486785 DOI: 10.1038/s41467-021-26031-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
Various behavioral and cognitive states exhibit circadian variations in animals across phyla including Drosophila melanogaster, in which only ~0.1% of the brain's neurons contain circadian clocks. Clock neurons transmit the timing information to a plethora of non-clock neurons via poorly understood mechanisms. Here, we address the molecular underpinning of this phenomenon by profiling circadian gene expression in non-clock neurons that constitute the mushroom body, the center of associative learning and sleep regulation. We show that circadian clocks drive rhythmic expression of hundreds of genes in mushroom body neurons, including the Neurofibromin 1 (Nf1) tumor suppressor gene and Pka-C1. Circadian clocks also drive calcium rhythms in mushroom body neurons via NF1-cAMP/PKA-C1 signaling, eliciting higher mushroom body activity during the day than at night, thereby promoting daytime wakefulness. These findings reveal the pervasive, non-cell-autonomous circadian regulation of gene expression in the brain and its role in sleep.
Collapse
Affiliation(s)
- Pedro Machado Almeida
- grid.8591.50000 0001 2322 4988Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva, 4, CH-1211 Switzerland
| | - Blanca Lago Solis
- grid.8591.50000 0001 2322 4988Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva, 4, CH-1211 Switzerland
| | - Luca Stickley
- grid.8591.50000 0001 2322 4988Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva, 4, CH-1211 Switzerland
| | - Alexis Feidler
- grid.8591.50000 0001 2322 4988Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva, 4, CH-1211 Switzerland ,grid.412750.50000 0004 1936 9166Present Address: University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Emi Nagoshi
- grid.8591.50000 0001 2322 4988Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva, 4, CH-1211 Switzerland
| |
Collapse
|
16
|
You S, Yu AM, Roberts MA, Joseph IJ, Jackson FR. Circadian regulation of the Drosophila astrocyte transcriptome. PLoS Genet 2021; 17:e1009790. [PMID: 34543266 PMCID: PMC8483315 DOI: 10.1371/journal.pgen.1009790] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/30/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Recent studies have demonstrated that astrocytes cooperate with neurons of the brain to mediate circadian control of many rhythmic processes including locomotor activity and sleep. Transcriptional profiling studies have described the overall rhythmic landscape of the brain, but few have employed approaches that reveal heterogeneous, cell-type specific rhythms of the brain. Using cell-specific isolation of ribosome-bound RNAs in Drosophila, we constructed the first circadian “translatome” for astrocytes. This analysis identified 293 “cycling genes” in astrocytes, most with mammalian orthologs. A subsequent behavioral genetic screen identified a number of genes whose expression is required in astrocytes for normal sleep behavior. In particular, we show that certain genes known to regulate fly innate immune responses are also required for normal sleep patterns.
Collapse
Affiliation(s)
- Samantha You
- Department of Neuroscience, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Alder M Yu
- Department of Biology, University of Wisconsin-La Crosse, La Crosse, Wisconsin, United States of America
| | - Mary A Roberts
- Department of Neuroscience, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ivanna J Joseph
- Department of Neuroscience, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - F Rob Jackson
- Department of Neuroscience, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
17
|
Huang Y, Lack JB, Hoppel GT, Pool JE. Parallel and Population-specific Gene Regulatory Evolution in Cold-Adapted Fly Populations. Genetics 2021; 218:6275754. [PMID: 33989401 PMCID: PMC8864734 DOI: 10.1093/genetics/iyab077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/10/2021] [Indexed: 11/15/2022] Open
Abstract
Changes in gene regulation at multiple levels may comprise an important share of the molecular changes underlying adaptive evolution in nature. However, few studies have assayed within- and between-population variation in gene regulatory traits at a transcriptomic scale, and therefore inferences about the characteristics of adaptive regulatory changes have been elusive. Here, we assess quantitative trait differentiation in gene expression levels and alternative splicing (intron usage) between three closely related pairs of natural populations of Drosophila melanogaster from contrasting thermal environments that reflect three separate instances of cold tolerance evolution. The cold-adapted populations were known to show population genetic evidence for parallel evolution at the SNP level, and here we find evidence for parallel expression evolution between them, with stronger parallelism at larval and adult stages than for pupae. We also implement a flexible method to estimate cis- vs trans-encoded contributions to expression or splicing differences at the adult stage. The apparent contributions of cis- vs trans-regulation to adaptive evolution vary substantially among population pairs. While two of three population pairs show a greater enrichment of cis-regulatory differences among adaptation candidates, trans-regulatory differences are more likely to be implicated in parallel expression changes between population pairs. Genes with significant cis-effects are enriched for signals of elevated genetic differentiation between cold- and warm-adapted populations, suggesting that they are potential targets of local adaptation. These findings expand our knowledge of adaptive gene regulatory evolution and our ability to make inferences about this important and widespread process.
Collapse
Affiliation(s)
- Yuheng Huang
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA.,Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Justin B Lack
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA.,Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Grant T Hoppel
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - John E Pool
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
18
|
Ma D, Przybylski D, Abruzzi KC, Schlichting M, Li Q, Long X, Rosbash M. A transcriptomic taxonomy of Drosophila circadian neurons around the clock. eLife 2021; 10:63056. [PMID: 33438579 PMCID: PMC7837698 DOI: 10.7554/elife.63056] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Many different functions are regulated by circadian rhythms, including those orchestrated by discrete clock neurons within animal brains. To comprehensively characterize and assign cell identity to the 75 pairs of Drosophila circadian neurons, we optimized a single-cell RNA sequencing method and assayed clock neuron gene expression at different times of day. The data identify at least 17 clock neuron categories with striking spatial regulation of gene expression. Transcription factor regulation is prominent and likely contributes to the robust circadian oscillation of many transcripts, including those that encode cell-surface proteins previously shown to be important for cell recognition and synapse formation during development. The many other clock-regulated genes also constitute an important resource for future mechanistic and functional studies between clock neurons and/or for temporal signaling to circuits elsewhere in the fly brain.
Collapse
Affiliation(s)
- Dingbang Ma
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Dariusz Przybylski
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Katharine C Abruzzi
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | | | - Qunlong Li
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Xi Long
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, United States
| | - Michael Rosbash
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| |
Collapse
|
19
|
Gunawardhana KL, Rivas GBS, Caster C, Hardin PE. Crosstalk between vrille transcripts, proteins, and regulatory elements controlling circadian rhythms and development in Drosophila. iScience 2020; 24:101893. [PMID: 33364582 PMCID: PMC7753146 DOI: 10.1016/j.isci.2020.101893] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/19/2020] [Accepted: 12/02/2020] [Indexed: 02/09/2023] Open
Abstract
The vrille (vri) gene encodes a transcriptional repressor required for Drosophila development as well as circadian behavior in adults. Alternate first exons produce vri transcripts predicted to produce a short VRI isoform during development and long VRI in adults. A vri mutant (vriΔ679) lacking long VRI transcripts is viable, confirming that short VRI is sufficient for developmental functions, yet behavioral rhythms in vriΔ679 flies persist, showing that short VRI is sufficient for clock output. E-box regulatory elements that drive rhythmic long VRI transcript expression are required for developmental expression of short VRI transcripts. Surprisingly, long VRI transcripts primarily produce short VRI in adults, apparently due to a poor Kozak sequence context, demonstrating that short VRI drives circadian behavior. Thus, E-box-driven long VRI transcripts primarily control circadian rhythms via short VRI, whereas the same E-boxes drive short VRI transcripts that control developmental functions using short VRI. vri-E mRNA is sufficient for Drosophila development and circadian behavior E-boxes upstream of the vri-ADF promoter are required for Drosophila development vri-ADF mRNAs primarily produce short VRI protein rather than long VRI protein Short VRI protein primarily controls Drosophila development and circadian behavior
Collapse
Affiliation(s)
- Kushan L Gunawardhana
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA
| | - Gustavo B S Rivas
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA
| | - Courtney Caster
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA
| | - Paul E Hardin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA
| |
Collapse
|
20
|
Kumar S, Tunc I, Tansey TR, Pirooznia M, Harbison ST. Identification of Genes Contributing to a Long Circadian Period in Drosophila Melanogaster. J Biol Rhythms 2020; 36:239-253. [PMID: 33274675 DOI: 10.1177/0748730420975946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endogenous circadian period of animals and humans is typically very close to 24 h. Individuals with much longer circadian periods have been observed, however, and in the case of humans, these deviations have health implications. Previously, we observed a line of Drosophila with a very long average period of 31.3 h for locomotor activity behavior. Preliminary mapping indicated that the long period did not map to known canonical clock genes but instead mapped to multiple chromosomes. Using RNA-Seq, we surveyed the whole transcriptome of fly heads from this line across time and compared it with a wild-type control. A three-way generalized linear model revealed that approximately two-thirds of the genes were expressed differentially among the two genotypes, while only one quarter of the genes varied across time. Using these results, we applied algorithms to search for genes that oscillated over 24 h, identifying genes not previously known to cycle. We identified 166 differentially expressed genes that overlapped with a previous Genome-wide Association Study (GWAS) of circadian behavior, strongly implicating them in the long-period phenotype. We tested mutations in 45 of these genes for their effect on the circadian period. Mutations in Alk, alph, CG10089, CG42540, CG6034, Kairos (CG6123), CG8768, klg, Lar, sick, and tinc had significant effects on the circadian period, with seven of these mutations increasing the circadian period of locomotor activity behavior. Genetic rescue of mutant Kairos restored the circadian period to wild-type levels, suggesting it has a critical role in determining period length in constant darkness.
Collapse
Affiliation(s)
- Shailesh Kumar
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Ilker Tunc
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Terry R Tansey
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Susan T Harbison
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| |
Collapse
|
21
|
Li Z, Rasmussen LJ. TIP60 in aging and neurodegeneration. Ageing Res Rev 2020; 64:101195. [PMID: 33091598 DOI: 10.1016/j.arr.2020.101195] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
Epigenetic modification of chromatin, including histone methylation and acetylation, plays critical roles in eukaryotic cells and has a significant impact on chromatin structure/accessibility, gene regulation and, susceptibility to aging, neurodegenerative disease, cancer, and other age-related diseases. This article reviews the current advances on TIP60/KAT5, a major histone acetyltransferase with diverse functions in eukaryotes, with emphasis on its regulation of autophagy, proteasome-dependent protein turnover, RNA transcription, DNA repair, circadian rhythms, learning and memory, and other neurological functions implicated in aging and neurodegeneration. Moreover, the promising therapeutic potential of TIP60 is discussed to target Alzheimer's disease and other neurological diseases.
Collapse
|
22
|
Wu G, Ruben MD, Lee Y, Li J, Hughes ME, Hogenesch JB. Genome-wide studies of time of day in the brain: Design and analysis. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.26599/bsa.2020.9050005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Transcriptome profiling at different times of day is powerful for studying circadian regulation in model organisms and humans. To date, 24 h profiles from many tissue types suggest that about half of all genes are circadian-expressed somewhere in the body. However, few of these studies focused on the brain. Thus, despite known links between circadian disruption and neurological disease, we have virtually no mechanistic understanding. In the coming decade, we expect more genome-wide studies of time of day in different brain diseases, regions, and cell types. We expect just as many different approaches to the design and analysis of these studies. This review considers key principles of circadian tran scriptomics, with the goal of maximizing utility and reproducibility of future studies in the nervous system.
Collapse
Affiliation(s)
- Gang Wu
- Divisions of Human Genetics and Immunobiology, Center for Chronobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH 45229, U.S.A
| | - Marc D. Ruben
- Divisions of Human Genetics and Immunobiology, Center for Chronobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH 45229, U.S.A
| | - Yinyeng Lee
- Divisions of Human Genetics and Immunobiology, Center for Chronobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH 45229, U.S.A
| | - Jiajia Li
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63310, U.S.A
| | - Michael E. Hughes
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63310, U.S.A
| | - John B. Hogenesch
- Divisions of Human Genetics and Immunobiology, Center for Chronobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH 45229, U.S.A
| |
Collapse
|
23
|
Zhang Z, So K, Peterson R, Bauer M, Ng H, Zhang Y, Kim JH, Kidd T, Miura P. Elav-Mediated Exon Skipping and Alternative Polyadenylation of the Dscam1 Gene Are Required for Axon Outgrowth. Cell Rep 2020; 27:3808-3817.e7. [PMID: 31242415 PMCID: PMC7092717 DOI: 10.1016/j.celrep.2019.05.083] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 04/18/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022] Open
Abstract
Many metazoan genes express alternative long 3′ UTR isoforms in the nervous system, but their functions remain largely unclear. In Drosophila melanogaster, the Dscam1 gene generates short and long (Dscam1-L) 3′ UTR isoforms because of alternative polyadenylation (APA). Here, we found that the RNA-binding protein Embryonic Lethal Abnormal Visual System (Elav) impacts Dscam1 biogenesis at two levels, including regulation of long 3′ UTR biogenesis and skipping of an upstream exon (exon 19). MinION long-read sequencing confirmed the connectivity of this alternative splicing event to the long 3′ UTR. Knockdown or CRISPR deletion of Dscam1-L impaired axon outgrowth in Drosophila. The Dscam1 long 3′ UTR was found to be required for correct Elav-mediated skipping of exon 19. Elav thus co-regulates APA and alternative splicing to generate specific Dscam1 transcripts that are essential for neural development. This coupling of APA to alternative splicing might represent a new class of regulated RNA processing. Like most metazoan genes, Dscam1 expresses alternative short and long 3′ UTR mRNAs. Zhang et al. find that loss of Dscam1 long 3′ UTR transcripts impairs axon outgrowth in Drosophila. Long-read sequencing reveals that these long 3′ UTR mRNAs preferentially skip an upstream exon, altering Dscam1 amino acid sequence.
Collapse
Affiliation(s)
- Zhiping Zhang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Kevin So
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Ryan Peterson
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Matthew Bauer
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Henry Ng
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Yong Zhang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Jung Hwan Kim
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Thomas Kidd
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Pedro Miura
- Department of Biology, University of Nevada, Reno, Reno, NV, USA.
| |
Collapse
|
24
|
Kozlov A, Koch R, Nagoshi E. Nitric oxide mediates neuro-glial interaction that shapes Drosophila circadian behavior. PLoS Genet 2020; 16:e1008312. [PMID: 32598344 PMCID: PMC7367490 DOI: 10.1371/journal.pgen.1008312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/17/2020] [Accepted: 06/05/2020] [Indexed: 11/25/2022] Open
Abstract
Drosophila circadian behavior relies on the network of heterogeneous groups of clock neurons. Short- and long-range signaling within the pacemaker circuit coordinates molecular and neural rhythms of clock neurons to generate coherent behavioral output. The neurochemistry of circadian behavior is complex and remains incompletely understood. Here we demonstrate that the gaseous messenger nitric oxide (NO) is a signaling molecule linking circadian pacemaker to rhythmic locomotor activity. We show that mutants lacking nitric oxide synthase (NOS) have behavioral arrhythmia in constant darkness, although molecular clocks in the main pacemaker neurons are unaffected. Behavioral phenotypes of mutants are due in part to the malformation of neurites of the main pacemaker neurons, s-LNvs. Using cell-type selective and stage-specific gain- and loss-of-function of NOS, we also demonstrate that NO secreted from diverse cellular clusters affect behavioral rhythms. Furthermore, we identify the perineurial glia, one of the two glial subtypes that form the blood-brain barrier, as the major source of NO that regulates circadian locomotor output. These results reveal for the first time the critical role of NO signaling in the Drosophila circadian system and highlight the importance of neuro-glial interaction in the neural circuit output. Circadian rhythms are daily cycles of physiological and behavioral processes found in most organisms on our planet from cyanobacteria to humans. Circadian rhythms allow organisms to anticipate routine daily and annual changes of environmental conditions and efficiently adapt to them. Fruit fly Drosophila melanogaster is an excellent model to study this phenomenon, as its versatile toolkit enables the study of genetic, molecular and neuronal mechanisms of rhythm generation. Here we report for the first time that gasotransmitter nitric oxide (NO) has a broad, multi-faceted impact on Drosophila circadian rhythms, which takes place both during the development and the adulthood. We also show that one of the important contributors of NO to circadian rhythms are glial cells that form the blood-brain barrier. The second finding highlights that circadian rhythms of higher organisms are not simply controlled by the small number of pacemaker neurons but are generated by the system that consists of many different players, including glia.
Collapse
Affiliation(s)
- Anatoly Kozlov
- Department of Genetics and Evolution, Sciences III, University of Geneva, Quai Ernest-Ansermet, Switzerland
| | - Rafael Koch
- Department of Genetics and Evolution, Sciences III, University of Geneva, Quai Ernest-Ansermet, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution, Sciences III, University of Geneva, Quai Ernest-Ansermet, Switzerland
- * E-mail:
| |
Collapse
|
25
|
Xu F, Kula-Eversole E, Iwanaszko M, Hutchison AL, Dinner A, Allada R. Circadian Clocks Function in Concert with Heat Shock Organizing Protein to Modulate Mutant Huntingtin Aggregation and Toxicity. Cell Rep 2020; 27:59-70.e4. [PMID: 30943415 PMCID: PMC7237104 DOI: 10.1016/j.celrep.2019.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/24/2019] [Accepted: 03/02/2019] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases commonly involve the disruption of circadian rhythms. Studies indicate that mutant Huntingtin (mHtt), the cause of Huntington’s disease (HD), disrupts circadian rhythms often before motor symptoms are evident. Yet little is known about the molecular mechanisms by which mHtt impairs circadian rhythmicity and whether circadian clocks can modulate HD pathogenesis. To address this question, we used a Drosophila HD model. We found that both environmental and genetic perturbations of the circadian clock alter mHtt-mediated neurodegeneration. To identify potential genetic pathways that mediate these effects, we applied a behavioral platform to screen for clock-regulated HD suppressors, identifying a role for Heat Shock Protein 70/90 Organizing Protein (Hop). Hop knockdown paradoxically reduces mHtt aggregation and toxicity. These studies demonstrate a role for the circadian clock in a neurodegenerative disease model and reveal a clock-regulated molecular and cellular pathway that links clock function to neurodegenerative disease. Disruption of circadian rhythms is frequently observed across a range of neurodegenerative diseases. Here, Xu et al. demonstrate that perturbation of circadian clocks alters the toxicity of the mutant Huntingtin protein, the cause of Huntington’s disease (HD). Moreover, they reveal a key mechanistic link between the clock and HD.
Collapse
Affiliation(s)
- Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | | | - Marta Iwanaszko
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alan L Hutchison
- Medical Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Aaron Dinner
- James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
26
|
Schlichting M. Entrainment of the Drosophila clock by the visual system. Neurosci Insights 2020; 15:2633105520903708. [PMID: 35174330 PMCID: PMC8842342 DOI: 10.1177/2633105520903708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/08/2020] [Indexed: 12/27/2022] Open
Abstract
Circadian clocks evolved as an adaptation to the cyclic change of day and night. To precisely adapt to this environment, the endogenous period has to be adjusted every day to exactly 24 hours by a process called entrainment. Organisms can use several external cues, called zeitgebers, to adapt. These include changes in temperature, humidity, or light. The latter is the most powerful signal to synchronize the clock in animals. Research shows that a complex visual system and circadian photoreceptors work together to adjust animal physiology to the outside world. This review will focus on the importance of the visual system for clock synchronization in the fruit fly Drosophila melanogaster. It will cover behavioral and physiological evidence that supports the importance of the visual system in light entrainment.
Collapse
|
27
|
Nair S, Bahn JH, Lee G, Yoo S, Park JH. A Homeobox Transcription Factor Scarecrow (SCRO) Negatively Regulates Pdf Neuropeptide Expression through Binding an Identified cis-Acting Element in Drosophila melanogaster. Mol Neurobiol 2020; 57:2115-2130. [DOI: 10.1007/s12035-020-01874-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
|
28
|
Agrawal P, Chung P, Heberlein U, Kent C. Enabling cell-type-specific behavioral epigenetics in Drosophila: a modified high-yield INTACT method reveals the impact of social environment on the epigenetic landscape in dopaminergic neurons. BMC Biol 2019; 17:30. [PMID: 30967153 PMCID: PMC6456965 DOI: 10.1186/s12915-019-0646-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/07/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Epigenetic mechanisms play fundamental roles in brain function and behavior and stressors such as social isolation can alter animal behavior via epigenetic mechanisms. However, due to cellular heterogeneity, identifying cell-type-specific epigenetic changes in the brain is challenging. Here, we report the first use of a modified isolation of nuclei tagged in specific cell type (INTACT) method in behavioral epigenetics of Drosophila melanogaster, a method we call mini-INTACT. RESULTS Using ChIP-seq on mini-INTACT purified dopaminergic nuclei, we identified epigenetic signatures in socially isolated and socially enriched Drosophila males. Social experience altered the epigenetic landscape in clusters of genes involved in transcription and neural function. Some of these alterations could be predicted by expression changes of four transcription factors and the prevalence of their binding sites in several clusters. These transcription factors were previously identified as activity-regulated genes, and their knockdown in dopaminergic neurons reduced the effects of social experience on sleep. CONCLUSIONS Our work enables the use of Drosophila as a model for cell-type-specific behavioral epigenetics and establishes that social environment shifts the epigenetic landscape in dopaminergic neurons. Four activity-related transcription factors are required in dopaminergic neurons for the effects of social environment on sleep.
Collapse
Affiliation(s)
- Pavan Agrawal
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Phuong Chung
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ulrike Heberlein
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Clement Kent
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Department of Biology, York University, Toronto, Canada.
| |
Collapse
|
29
|
Aguglia A, Serafini G, Solano P, Giacomini G, Conigliaro C, Salvi V, Mencacci C, Romano M, Aguglia E, Amore M. The role of seasonality and photoperiod on the lethality of suicide attempts: A case-control study. J Affect Disord 2019; 246:895-901. [PMID: 30795496 DOI: 10.1016/j.jad.2018.12.094] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/19/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND The risk factors related to suicidal behaviors are complex and not yet fully known. Several studies underline how suicide results from the combination of psycho-social, biological, cultural, and environmental factors. The aim of this study was to investigate the potential role of seasonality and photoperiod on high-lethality suicide attempts (HLSA) compared with low-lethality suicide attempts (LLSA) in a sample of psychiatric inpatients. METHODS After attempting suicide, subjects were admitted in the emergency/psychiatric ward of the IRCCS Ospedale Policlinico San Martino from 1st August 2013 to 31st July 2018. Socio-demographic and clinical characteristics were collected. RESULTS The sample consisted of four hundred thirty-two individuals admitted for suicide attempt. One hundred thirty-three subjects (30.8%) of the sample committed a HLSA. The HLSA group peaked in the months with a higher sunlight exposure (June and July). Bivariate correlation analyses between seasonality/photoperiod in the whole sample and HLSA were positively associated with summer and highest solar intensity period. LIMITATIONS Data were limited to a single hospital, patients' seasonal environment, meteorological variables and psychological factors. In addition, the presence of acute life-events fostering the suicidal crisis has not been investigated. CONCLUSIONS The current study provides a novel perspective on the questions surrounding the impact of seasonality and daylight exposure on lethality of suicide attempts. further studies are needed to provide deeper understandings on the delicate molecular network that links suicide behaviors, seasonality and daylight in order to develop more effective prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Andrea Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Solano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gabriele Giacomini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Conigliaro
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Virginio Salvi
- Department of Neuroscience, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Claudio Mencacci
- Department of Neuroscience, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Miroslav Romano
- Department of Experimental and Clinical Medicine, Psychiatric Clinic University Hospital "Gaspare Rodolico", University of Catania, Catania, Italy
| | - Eugenio Aguglia
- Department of Experimental and Clinical Medicine, Psychiatric Clinic University Hospital "Gaspare Rodolico", University of Catania, Catania, Italy
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics Maternal and Child Health, University of Genoa, Section of Psychiatry, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
30
|
Abstract
A fundamental question in contemporary neuroscience is how the remarkable cellular diversity required for the intricate function of the nervous system is achieved. Here, we bridge the gap between a cellular machinery that is known to diversify the transcriptome and the existence of distinct neuronal populations that compose the Drosophila brain. Adenosine-to-inosine (A-to-I) RNA editing is a ubiquitous mechanism that generates transcriptomic diversity in cells by recoding certain adenosines within the pre-mRNA sequence into inosines. We present a spatial map of RNA editing across different neuronal populations in Drosophila brain. Each neuronal population has a distinct editing signature, with the majority of differential editing occurring in highly conserved regions of transcripts that encode ion channels and other essential neuronal genes. Adenosine-to-inosine (A-to-I) RNA editing, catalyzed by ADAR enzymes, is a ubiquitous mechanism that generates transcriptomic diversity. This process is particularly important for proper neuronal function; however, little is known about how RNA editing is dynamically regulated between the many functionally distinct neuronal populations of the brain. Here, we present a spatial RNA editing map in the Drosophila brain and show that different neuronal populations possess distinct RNA editing signatures. After purifying and sequencing RNA from genetically marked groups of neuronal nuclei, we identified a large number of editing sites and compared editing levels in hundreds of transcripts across nine functionally different neuronal populations. We found distinct editing repertoires for each population, including sites in repeat regions of the transcriptome and differential editing in highly conserved and likely functional regions of transcripts that encode essential neuronal genes. These changes are site-specific and not driven by changes in Adar expression, suggesting a complex, targeted regulation of editing levels in key transcripts. This fine-tuning of the transcriptome between different neurons by RNA editing may account for functional differences between distinct populations in the brain.
Collapse
|
31
|
Du J, Zhang Y, Xue Y, Zhao X, Zhao X, Wei Y, Li Z, Zhang Y, Zhao Z. Diurnal protein oscillation profiles in Drosophila head. FEBS Lett 2018; 592:3736-3749. [PMID: 30311939 DOI: 10.1002/1873-3468.13267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/23/2018] [Accepted: 10/02/2018] [Indexed: 11/08/2022]
Abstract
Circadian clocks control daily rhythms in physiology, metabolism and behaviour in most organisms. Proteome-wide analysis of protein oscillations is still lacking in Drosophila. In this study, the total protein and phosphorylated protein in Drosophila heads in a 24-h daily time-course were assayed by using the isobaric tags for relative and absolute quantitation (iTRAQ) method, and 10 and 7 oscillating proteins as well as 19 and 22 oscillating phosphoproteins in the w1118 control and ClkJrk mutant strains were separately identified. Lastly, we performed a mini screen to investigate the functions of some oscillating proteins in circadian locomotion rhythms. This study provides the first proteomic profiling of diurnally oscillating proteins in fly heads, thereby providing a basis for further mechanistic studies of these proteins in circadian rhythm.
Collapse
Affiliation(s)
- Juan Du
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yifan Zhang
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yongbo Xue
- Department of Biology, University of Nevada, Reno, NV, USA
| | - Xiaoyun Zhao
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xianguo Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yu Wei
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhen Li
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yong Zhang
- Department of Biology, University of Nevada, Reno, NV, USA
| | - Zhangwu Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
32
|
Drosophila Models of Sporadic Parkinson's Disease. Int J Mol Sci 2018; 19:ijms19113343. [PMID: 30373150 PMCID: PMC6275057 DOI: 10.3390/ijms19113343] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is the most common cause of movement disorders and is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. It is increasingly recognized as a complex group of disorders presenting widely heterogeneous symptoms and pathology. With the exception of the rare monogenic forms, the majority of PD cases result from an interaction between multiple genetic and environmental risk factors. The search for these risk factors and the development of preclinical animal models are in progress, aiming to provide mechanistic insights into the pathogenesis of PD. This review summarizes the studies that capitalize on modeling sporadic (i.e., nonfamilial) PD using Drosophilamelanogaster and discusses their methodologies, new findings, and future perspectives.
Collapse
|
33
|
Hughes KL, Abshire ET, Goldstrohm AC. Regulatory roles of vertebrate Nocturnin: insights and remaining mysteries. RNA Biol 2018; 15:1255-1267. [PMID: 30257600 PMCID: PMC6284557 DOI: 10.1080/15476286.2018.1526541] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022] Open
Abstract
Post-transcriptional control of messenger RNA (mRNA) is an important layer of gene regulation that modulates mRNA decay, translation, and localization. Eukaryotic mRNA decay begins with the catalytic removal of the 3' poly-adenosine tail by deadenylase enzymes. Multiple deadenylases have been identified in vertebrates and are known to have distinct biological roles; among these proteins is Nocturnin, which has been linked to circadian biology, adipogenesis, osteogenesis, and obesity. Multiple studies have investigated Nocturnin's involvement in these processes; however, a full understanding of its molecular function remains elusive. Recent studies have provided new insights by identifying putative Nocturnin-regulated mRNAs in mice and by determining the structure and regulatory activities of human Nocturnin. This review seeks to integrate these new discoveries into our understanding of Nocturnin's regulatory functions and highlight the important remaining unanswered questions surrounding its regulation, biochemical activities, protein partners, and target mRNAs.
Collapse
Affiliation(s)
- Kelsey L. Hughes
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Elizabeth T. Abshire
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Aaron C. Goldstrohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
NonA and CPX Link the Circadian Clockwork to Locomotor Activity in Drosophila. Neuron 2018; 99:768-780.e3. [PMID: 30057203 DOI: 10.1016/j.neuron.2018.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/27/2018] [Accepted: 07/01/2018] [Indexed: 11/20/2022]
Abstract
Drosophila NonA and its mammalian ortholog NONO are members of the Drosophila behavior and human splicing (DBHS) family. NONO also has a strong circadian connection: it associates with the circadian repressor protein PERIOD (PER) and contributes to circadian timekeeping. Here, we investigate NonA, which is required for proper levels of evening locomotor activity as well as a normal free-running period in Drosophila. NonA is associated with the positive transcription factor CLOCK/CYCLE (CLK/CYC), interacts directly with complexin (cpx) pre-mRNA, and upregulates gene expression, including the gene cpx. Downregulation of cpx expression in circadian neurons phenocopies NonA downregulation, whereas cpx overexpression rescues the nonA RNAi phenotypes, indicating that cpx is an important NonA target gene. As the cpx protein contributes to proper neurotransmitter and neuropeptide release in response to calcium, these results and others indicate that this control is important for the normal circadian regulation of locomotor activity.
Collapse
|
35
|
Top D, Young MW. Coordination between Differentially Regulated Circadian Clocks Generates Rhythmic Behavior. Cold Spring Harb Perspect Biol 2018; 10:a033589. [PMID: 28893860 PMCID: PMC6028074 DOI: 10.1101/cshperspect.a033589] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Specialized groups of neurons in the brain are key mediators of circadian rhythms, receiving daily environmental cues and communicating those signals to other tissues in the organism for entrainment and to organize circadian physiology. In Drosophila, the "circadian clock" is housed in seven neuronal clusters, which are defined by their expression of the main circadian proteins, Period, Timeless, Clock, and Cycle. These clusters are distributed across the fly brain and are thereby subject to the respective environments associated with their anatomical locations. While these core components are universally expressed in all neurons of the circadian network, additional regulatory proteins that act on these components are differentially expressed, giving rise to "local clocks" within the network that nonetheless converge to regulate coherent behavioral rhythms. In this review, we describe the communication between the neurons of the circadian network and the molecular differences within neurons of this network. We focus on differences in protein-expression patterns and discuss how such variation can impart functional differences in each local clock. Finally, we summarize our current understanding of how communication within the circadian network intersects with intracellular biochemical mechanisms to ultimately specify behavioral rhythms. We propose that additional efforts are required to identify regulatory mechanisms within each neuronal cluster to understand the molecular basis of circadian behavior.
Collapse
Affiliation(s)
- Deniz Top
- Laboratory of Genetics, The Rockefeller University, New York, New York 10065
| | - Michael W Young
- Laboratory of Genetics, The Rockefeller University, New York, New York 10065
| |
Collapse
|
36
|
Wang Q, Abruzzi KC, Rosbash M, Rio DC. Striking circadian neuron diversity and cycling of Drosophila alternative splicing. eLife 2018; 7:35618. [PMID: 29863472 PMCID: PMC6025963 DOI: 10.7554/elife.35618] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/31/2018] [Indexed: 11/13/2022] Open
Abstract
Although alternative pre-mRNA splicing (AS) significantly diversifies the neuronal proteome, the extent of AS is still unknown due in part to the large number of diverse cell types in the brain. To address this complexity issue, we used an annotation-free computational method to analyze and compare the AS profiles between small specific groups of Drosophila circadian neurons. The method, the Junction Usage Model (JUM), allows the comprehensive profiling of both known and novel AS events from specific RNA-seq libraries. The results show that many diverse and novel pre-mRNA isoforms are preferentially expressed in one class of clock neuron and also absent from the more standard Drosophila head RNA preparation. These AS events are enriched in potassium channels important for neuronal firing, and there are also cycling isoforms with no detectable underlying transcriptional oscillations. The results suggest massive AS regulation in the brain that is also likely important for circadian regulation.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Center for RNA Systems Biology (CRSB), University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, United States
| | - Katharine C Abruzzi
- Department of Biology, Howard Hughes Medical Institute, Brandeis University, Waltham, United States.,National Center for Behavior Genomics, Brandeis University, Waltham, United States
| | - Michael Rosbash
- Department of Biology, Howard Hughes Medical Institute, Brandeis University, Waltham, United States.,National Center for Behavior Genomics, Brandeis University, Waltham, United States
| | - Donald C Rio
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Center for RNA Systems Biology (CRSB), University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, United States
| |
Collapse
|
37
|
Croset V, Treiber CD, Waddell S. Cellular diversity in the Drosophila midbrain revealed by single-cell transcriptomics. eLife 2018; 7:34550. [PMID: 29671739 PMCID: PMC5927767 DOI: 10.7554/elife.34550] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
To understand the brain, molecular details need to be overlaid onto neural wiring diagrams so that synaptic mode, neuromodulation and critical signaling operations can be considered. Single-cell transcriptomics provide a unique opportunity to collect this information. Here we present an initial analysis of thousands of individual cells from Drosophila midbrain, that were acquired using Drop-Seq. A number of approaches permitted the assignment of transcriptional profiles to several major brain regions and cell-types. Expression of biosynthetic enzymes and reuptake mechanisms allows all the neurons to be typed according to the neurotransmitter or neuromodulator that they produce and presumably release. Some neuropeptides are preferentially co-expressed in neurons using a particular fast-acting transmitter, or monoamine. Neuromodulatory and neurotransmitter receptor subunit expression illustrates the potential of these molecules in generating complexity in neural circuit function. This cell atlas dataset provides an important resource to link molecular operations to brain regions and complex neural processes.
Collapse
Affiliation(s)
- Vincent Croset
- Centre for Neural Circuits and Behaviour, The University of Oxford, Oxford, United Kingdom
| | - Christoph D Treiber
- Centre for Neural Circuits and Behaviour, The University of Oxford, Oxford, United Kingdom
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, The University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Widmann A, Eichler K, Selcho M, Thum AS, Pauls D. Odor-taste learning in Drosophila larvae. JOURNAL OF INSECT PHYSIOLOGY 2018; 106:47-54. [PMID: 28823531 DOI: 10.1016/j.jinsphys.2017.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/07/2017] [Accepted: 08/16/2017] [Indexed: 06/07/2023]
Abstract
The Drosophila larva is an attractive model system to study fundamental questions in the field of neuroscience. Like the adult fly, the larva offers a seemingly unlimited genetic toolbox, which allows one to visualize, silence or activate neurons down to the single cell level. This, combined with its simplicity in terms of cell numbers, offers a useful system to study the neuronal correlates of complex processes including associative odor-taste learning and memory formation. Here, we summarize the current knowledge about odor-taste learning and memory at the behavioral level and integrate the recent progress on the larval connectome to shed light on the sub-circuits that allow Drosophila larvae to integrate present sensory input in the context of past experience and to elicit an appropriate behavioral response.
Collapse
Affiliation(s)
| | - Katharina Eichler
- Department of Biology, University of Konstanz, D-78464 Konstanz, Germany; HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Mareike Selcho
- Department of Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany
| | - Andreas S Thum
- Department of Biology, University of Konstanz, D-78464 Konstanz, Germany; Department of Genetics, University of Leipzig, D-04103 Leipzig, Germany.
| | - Dennis Pauls
- Department of Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany.
| |
Collapse
|
39
|
Tas D, Stickley L, Miozzo F, Koch R, Loncle N, Sabado V, Gnägi B, Nagoshi E. Parallel roles of transcription factors dFOXO and FER2 in the development and maintenance of dopaminergic neurons. PLoS Genet 2018. [PMID: 29529025 PMCID: PMC5864087 DOI: 10.1371/journal.pgen.1007271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Forkhead box (FOXO) proteins are evolutionarily conserved, stress-responsive transcription factors (TFs) that can promote or counteract cell death. Mutations in FOXO genes are implicated in numerous pathologies, including age-dependent neurodegenerative disorders, such as Parkinson’s disease (PD). However, the complex regulation and downstream mechanisms of FOXOs present a challenge in understanding their roles in the pathogenesis of PD. Here, we investigate the involvement of FOXO in the death of dopaminergic (DA) neurons, the key pathological feature of PD, in Drosophila. We show that dFOXO null mutants exhibit a selective loss of DA neurons in the subgroup crucial for locomotion, the protocerebral anterior medial (PAM) cluster, during development as well as in adulthood. PAM neuron-targeted adult-restricted knockdown demonstrates that dFOXO in adult PAM neurons tissue-autonomously promotes neuronal survival during aging. We further show that dFOXO and the bHLH-TF 48-related-2 (FER2) act in parallel to protect PAM neurons from different forms of cellular stress. Remarkably, however, dFOXO and FER2 share common downstream processes leading to the regulation of autophagy and mitochondrial morphology. Thus, overexpression of one can rescue the loss of function of the other. These results indicate a role of dFOXO in neuroprotection and highlight the notion that multiple genetic and environmental factors interact to increase the risk of DA neuron degeneration and the development of PD. PD, mainly characterized by a progressive loss of dopaminergic neurons in the substantia nigra (SN), is the most prevalent neurodegenerative movement disorder affecting more than 6 million people worldwide. Despite the discovery of several genes linked to familial PD, our understanding of its pathogenesis remains limited, as approximately 90% of the PD cases are sporadic with no apparent genetic linkage. Genome-wide expression studies have implicated the stress-responsive TF FOXO in PD. However, the exact role of FOXO in the survival of DA neurons and PD pathogenesis is still poorly understood. Here, we use fruit flies to address the role of FOXO in the maintenance of DA neurons. dFOXO (Drosophila FOXO) null mutants show a progressive loss of DA neurons in the subgroup essential for locomotion, a phenotype identical to that of Fer2 mutants. Remarkably, dFOXO and FER2 act in parallel pathways to protect PAM neurons from different cellular stressors, but both pathways contribute to the regulation of autophagy and mitochondrial biology. These results demonstrate that dFOXO is required for the maintenance of DA neurons important for locomotion and shed new light on the molecular mechanisms underpinning the complex gene-environment interactions affecting DA neuron survival and PD pathogenesis.
Collapse
Affiliation(s)
- Damla Tas
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Luca Stickley
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Federico Miozzo
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Rafael Koch
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Nicolas Loncle
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Virginie Sabado
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
| | - Bettina Gnägi
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, Bern, CH, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution, Sciences III, University of Geneva, 30 Quai Ernest-Ansermet, Geneva-4, CH, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
40
|
Mezan S, Feuz JD, Deplancke B, Kadener S. PDF Signaling Is an Integral Part of the Drosophila Circadian Molecular Oscillator. Cell Rep 2017; 17:708-719. [PMID: 27732848 PMCID: PMC5081397 DOI: 10.1016/j.celrep.2016.09.048] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 08/12/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022] Open
Abstract
Circadian clocks generate 24-hr rhythms in physiology and behavior. Despite numerous studies, it is still uncertain how circadian rhythms emerge from their molecular and neural constituents. Here, we demonstrate a tight connection between the molecular and neuronal circadian networks. Using fluorescent transcriptional reporters in a Drosophila ex vivo brain culture system, we identified a reciprocal negative regulation between the master circadian regulator CLK and expression of pdf, the main circadian neuropeptide. We show that PDF feedback is required for maintaining normal oscillation pattern in CLK-driven transcription. Interestingly, we found that CLK and neuronal firing suppresses pdf transcription, likely through a common pathway involving the transcription factors DHR38 and SR, establishing a direct link between electric activity and the circadian system. In sum, our work provides evidence for the existence of an uncharacterized CLK-PDF feedback loop that tightly wraps together the molecular oscillator with the circadian neuronal network in Drosophila. Monitoring circadian transcription ex vivo using fluorescent reporters CLK activation in the LNvs provokes downregulation in CLK activity in LNds and DNs Reciprocal negative regulation of CLK activity and pdf transcription and signaling PDF signaling is required for the normal oscillation pattern in CLK activity
Collapse
Affiliation(s)
- Shaul Mezan
- Biological Chemistry Department, Silberman Institute of Life Sciences, the Hebrew University, Jerusalem 91904, Israel
| | - Jean Daniel Feuz
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Bart Deplancke
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Sebastian Kadener
- Biological Chemistry Department, Silberman Institute of Life Sciences, the Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
41
|
Adewoye AB, Nuzhdin SV, Tauber E. Mapping Quantitative Trait Loci Underlying Circadian Light Sensitivity in Drosophila. J Biol Rhythms 2017; 32:394-405. [PMID: 28990443 DOI: 10.1177/0748730417731863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Despite the significant advance in our understanding of the molecular basis of light entrainment of the circadian clock in Drosophila, the underlying genetic architecture is still largely unknown. The aim of this study was to identify loci associated with variation in circadian photosensitivity, which are important for the evolution of this trait. We have used complementary approaches that combined quantitative trait loci (QTL) mapping, complementation testing, and transcriptome profiling to dissect this variation. We identified a major QTL on chromosome 2, which was subsequently fine mapped using deficiency complementation mapping into 2 smaller regions spanning 139 genes, some of which are known to be involved in functions that have been previously implicated in light entrainment. Two genes implicated with the clock and located within that interval, timeless and cycle, failed to complement the QTL, indicating that alleles of these genes contribute to the variation in light response. Specifically, we find that the timeless s/ ls polymorphism that has been previously shown to constitute a latitudinal cline in Europe is also segregating in our recombinant inbred lines and is contributing to the phenotypic variation in light sensitivity. We also profiled gene expression in 2 recombinant inbred strains that differ significantly in their photosensitivity and identified a total of 368 transcripts that showed differential expression (false discovery rate < 0.1). Of 131 transcripts that showed a significant recombinant inbred line by treatment interaction (i.e., putative expression QTL), 4 are located within QTL2.
Collapse
Affiliation(s)
- Adeolu B Adewoye
- Department of Genetics, University of Leicester, Leicester, UK.,1 Wolfson School of Mechanical and Manufacturing Engineering, Centre for Biological Engineering, Loughborough University Loughborough, UK
| | - Sergey V Nuzhdin
- Program in Molecular and Computation Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California, USA
| | - Eran Tauber
- Department of Genetics, University of Leicester, Leicester, UK.,Department of Evolutionary and Environmental Biology and Institute of Evolution, University of Haifa, Haifa, Israel
| |
Collapse
|
42
|
Abstract
The Drosophila circadian clock keeps time via transcriptional feedback loops. These feedback loops are initiated by CLOCK-CYCLE (CLK-CYC) heterodimers, which activate transcription of genes encoding the feedback repressors PERIOD and TIMELESS. Circadian clocks normally operate in ∼150 brain pacemaker neurons and in many peripheral tissues in the head and body, but can also be induced by expressing CLK in nonclock cells. These ectopic clocks also require cyc, yet CYC expression is restricted to canonical clock cells despite evidence that cyc mRNA is widely expressed. Here we show that CLK binds to and stabilizes CYC in cell culture and in nonclock cells in vivo. Ectopic clocks also require the blue light photoreceptor CRYPTOCHROME (CRY), which is required for both light entrainment and clock function in peripheral tissues. These experiments define the genetic architecture required to initiate circadian clock function in Drosophila, reveal mechanisms governing circadian activator stability that are conserved in perhaps all eukaryotes, and suggest that Clk, cyc, and cry expression is sufficient to drive clock expression in naive cells.
Collapse
|
43
|
Hou Z, Su L, Pei J, Grishin NV, Zhang H. Crystal Structure of the CLOCK Transactivation Domain Exon19 in Complex with a Repressor. Structure 2017; 25:1187-1194.e3. [PMID: 28669630 DOI: 10.1016/j.str.2017.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/04/2017] [Accepted: 05/25/2017] [Indexed: 11/24/2022]
Abstract
In the canonical clock model, CLOCK:BMAL1-mediated transcriptional activation is feedback regulated by its repressors CRY and PER and, in association with other coregulators, ultimately generates oscillatory gene expression patterns. How CLOCK:BMAL1 interacts with coregulator(s) is not well understood. Here we report the crystal structures of the mouse CLOCK transactivating domain Exon19 in complex with CIPC, a potent circadian repressor that functions independently of CRY and PER. The Exon19:CIPC complex adopts a three-helical coiled-coil bundle conformation containing two Exon19 helices and one CIPC. Unique to Exon19:CIPC, three highly conserved polar residues, Asn341 of CIPC and Gln544 of the two Exon19 helices, are located at the mid-section of the coiled-coil bundle interior and form hydrogen bonds with each other. Combining results from protein database search, sequence analysis, and mutagenesis studies, we discovered for the first time that CLOCK Exon19:CIPC interaction is a conserved transcription regulatory mechanism among mammals, fish, flies, and other invertebrates.
Collapse
Affiliation(s)
- Zhiqiang Hou
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lijing Su
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jimin Pei
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hong Zhang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
44
|
Treiber CD, Waddell S. Resolving the prevalence of somatic transposition in Drosophila. eLife 2017; 6. [PMID: 28742021 PMCID: PMC5553932 DOI: 10.7554/elife.28297] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/21/2017] [Indexed: 11/13/2022] Open
Abstract
Somatic transposition in mammals and insects could increase cellular diversity and neural mobilization has been implicated in age-dependent decline. To understand the impact of transposition in somatic cells it is essential to reliably measure the frequency and map locations of new insertions. Here we identified thousands of putative somatic transposon insertions in neurons from individual Drosophila melanogaster using whole-genome sequencing. However, the number of de novo insertions did not correlate with transposon expression or fly age. Analysing our data with exons as 'immobile genetic elements' revealed a similar frequency of unexpected exon translocations. A new sequencing strategy that recovers transposon: chromosome junction information revealed most putative de novo transposon and exon insertions likely result from unavoidable chimeric artefacts. Reanalysis of other published data suggests similar artefacts are often mistaken for genuine somatic transposition. We conclude that somatic transposition is less prevalent in Drosophila than previously envisaged.
Collapse
Affiliation(s)
- Christoph D Treiber
- Centre for Neural Circuits and Behaviour, The University of Oxford, Oxford, United Kingdom
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, The University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
A Screening of UNF Targets Identifies Rnb, a Novel Regulator of Drosophila Circadian Rhythms. J Neurosci 2017; 37:6673-6685. [PMID: 28592698 DOI: 10.1523/jneurosci.3286-16.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 11/21/2022] Open
Abstract
Behavioral circadian rhythms are controlled by multioscillator networks comprising functionally different subgroups of clock neurons. Studies have demonstrated that molecular clocks in the fruit fly Drosophila melanogaster are regulated differently in clock neuron subclasses to support their specific functions (Lee et al., 2016; Top et al., 2016). The nuclear receptor unfulfilled (unf) represents a regulatory node that provides the small ventral lateral neurons (s-LNvs) unique characteristics as the master pacemaker (Beuchle et al., 2012). We previously showed that UNF interacts with the s-LNv molecular clocks by regulating transcription of the core clock gene period (per) (Jaumouillé et al., 2015). To gain more insight into the mechanisms by which UNF contributes to the functioning of the circadian master pacemaker, we identified UNF target genes using chromatin immunoprecipitation. Our data demonstrate that a previously uncharacterized gene CG7837, which we termed R and B (Rnb), acts downstream of UNF to regulate the function of the s-LNvs as the master circadian pacemaker. Mutations and LNv-targeted adult-restricted knockdown of Rnb impair locomotor rhythms. RNB localizes to the nucleus, and its loss-of-function blunts the molecular rhythms and output rhythms of the s-LNvs, particularly the circadian rhythms in PDF accumulation and axonal arbor remodeling. These results establish a second pathway by which UNF interacts with the molecular clocks in the s-LNvs and highlight the mechanistic differences in the molecular clockwork within the pacemaker circuit.SIGNIFICANCE STATEMENT Circadian behavior is generated by a pacemaker circuit comprising diverse classes of pacemaker neurons, each of which contains a molecular clock. In addition to the anatomical and functional diversity, recent studies have shown the mechanistic differences in the molecular clockwork among the pacemaker neurons in Drosophila Here, we identified the molecular characteristics distinguishing the s-LNvs, the master pacemaker of the locomotor rhythms, from other clock neuron subtypes. We demonstrated that a newly identified gene Rnb is an s-LNv-specific regulator of the molecular clock and essential for the generation of circadian locomotor behavior. Our results provide additional evidence to the emerging view that the differential regulation of the molecular clocks underlies the functional differences among the pacemaker neuron subgroups.
Collapse
|
46
|
Cardinal Epigenetic Role of non-coding Regulatory RNAs in Circadian Rhythm. Mol Neurobiol 2017; 55:3564-3576. [PMID: 28516429 DOI: 10.1007/s12035-017-0573-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
Circadian rhythm which governs basic physiological activities like sleeping, feeding and energy consumption is regulated by light-controlled central clock genes in the pacemaker neuron. The timekeeping machinery with unique transcriptional and post-transcriptional feedback loops is controlled by different small regulatory RNAs in the brain. Roles of the multiple neuronal genes, especially post-transcriptional regulation, splicing, polyadenylation, mature mRNA editing, and stability of translation products, are controlled by epigenetic activities orchestrated via small RNAs. Collectively, these mechanisms regulate clock and light-controlled genes for effecting pacemaker activity and entrainment. Regulatory small RNAs of the circadian circuit, timekeeping mechanism, synchronization of regular entrainment, oscillation, and rhythmicity are regulated by diversified RNA molecules. Regulatory small RNAs operate critical roles in brain activities including the neuronal clock activity. In this report, we propose the emergence of the earlier unexpected small RNAs for a historic perspective of epigenetic regulation of the brain clock system.
Collapse
|
47
|
MicroRNA-92a is a circadian modulator of neuronal excitability in Drosophila. Nat Commun 2017; 8:14707. [PMID: 28276426 PMCID: PMC5347142 DOI: 10.1038/ncomms14707] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/24/2017] [Indexed: 01/01/2023] Open
Abstract
Many biological and behavioural processes of animals are governed by an endogenous circadian clock, which is dependent on transcriptional regulation. Here we address post-transcriptional regulation and the role of miRNAs in Drosophila circadian rhythms. At least six miRNAs show cycling expression levels within the pigment dispersing factor (PDF) cell-pacemaker neurons; only mir-92a peaks during the night. In vivo calcium monitoring, dynamics of PDF projections, ArcLight, GCaMP6 imaging and sleep assays indicate that mir-92a suppresses neuronal excitability. In addition, mir-92a levels within PDF cells respond to light pulses and also affect the phase shift response. Translating ribosome affinity purification (TRAP) and in vitro luciferase reporter assay indicate that mir-92a suppresses expression of sirt2, which is homologous to human sir2 and sirt3. sirt2 RNAi also phenocopies mir-92a overexpression. These experiments indicate that sirt2 is a functional mir-92a target and that mir-92a modulates PDF neuronal excitability via suppressing SIRT2 levels in a rhythmic manner. Accumulating evidence suggests that microRNAs play a role in circadian regulation. Here the authors show that in the Drosophila brain, mir-92a suppresses the excitability of PDF neurons—key circadian pacemaker cells in Drosophila—via inhibiting the translation of its target sirt2.
Collapse
|
48
|
Abruzzi KC, Zadina A, Luo W, Wiyanto E, Rahman R, Guo F, Shafer O, Rosbash M. RNA-seq analysis of Drosophila clock and non-clock neurons reveals neuron-specific cycling and novel candidate neuropeptides. PLoS Genet 2017; 13:e1006613. [PMID: 28182648 PMCID: PMC5325595 DOI: 10.1371/journal.pgen.1006613] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/24/2017] [Accepted: 02/01/2017] [Indexed: 12/21/2022] Open
Abstract
Locomotor activity rhythms are controlled by a network of ~150 circadian neurons within the adult Drosophila brain. They are subdivided based on their anatomical locations and properties. We profiled transcripts “around the clock” from three key groups of circadian neurons with different functions. We also profiled a non-circadian outgroup, dopaminergic (TH) neurons. They have cycling transcripts but fewer than clock neurons as well as low expression and poor cycling of clock gene transcripts. This suggests that TH neurons do not have a canonical circadian clock and that their gene expression cycling is driven by brain systemic cues. The three circadian groups are surprisingly diverse in their cycling transcripts and overall gene expression patterns, which include known and putative novel neuropeptides. Even the overall phase distributions of cycling transcripts are distinct, indicating that different regulatory principles govern transcript oscillations. This surprising cell-type diversity parallels the functional heterogeneity of the different neurons. Organisms ranging from bacteria to humans contain circadian clocks. They keep internal time and also integrate environmental cues such as light to provide external time information for entrainment. In the fruit fly Drosophila melanogaster, ~150 brain neurons contain the circadian machinery and are critical for controlling behavior. Several subgroups of these clock neurons have been identified by their anatomical locations and specific functions. Our work aims to profile these neurons and to characterize their molecular contents: what to they contain and how do they differ? To this end, we have purified 3 important subgroups of clock neurons and identified their expressed genes at different times of day. Some are expressed at all times, whereas others are “cycling,” i.e., expressed more strongly at a particular time of day like the morning. Interestingly, each circadian subgroup is quite different. The data provide hints about what functions each group of neurons carries out and how they may work together to keep time. In addition, even a non-circadian group of neurons has cycling genes and has implications for the extent to which all cells have or do not have a functional circadian clock.
Collapse
Affiliation(s)
- Katharine C. Abruzzi
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Abigail Zadina
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Weifei Luo
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Evelyn Wiyanto
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Reazur Rahman
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Fang Guo
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Orie Shafer
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Michael Rosbash
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
- * E-mail:
| |
Collapse
|
49
|
Millius A, Ueda HR. Systems Biology-Derived Discoveries of Intrinsic Clocks. Front Neurol 2017; 8:25. [PMID: 28220104 PMCID: PMC5292584 DOI: 10.3389/fneur.2017.00025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/17/2017] [Indexed: 12/19/2022] Open
Abstract
A systems approach to studying biology uses a variety of mathematical, computational, and engineering tools to holistically understand and model properties of cells, tissues, and organisms. Building from early biochemical, genetic, and physiological studies, systems biology became established through the development of genome-wide methods, high-throughput procedures, modern computational processing power, and bioinformatics. Here, we highlight a variety of systems approaches to the study of biological rhythms that occur with a 24-h period-circadian rhythms. We review how systems methods have helped to elucidate complex behaviors of the circadian clock including temperature compensation, rhythmicity, and robustness. Finally, we explain the contribution of systems biology to the transcription-translation feedback loop and posttranslational oscillator models of circadian rhythms and describe new technologies and "-omics" approaches to understand circadian timekeeping and neurophysiology.
Collapse
Affiliation(s)
- Arthur Millius
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka, Japan
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Pankova K, Borst A. Transgenic line for the identification of cholinergic release sites in Drosophila melanogaster. ACTA ACUST UNITED AC 2017; 220:1405-1410. [PMID: 28167805 PMCID: PMC5413067 DOI: 10.1242/jeb.149369] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/31/2017] [Indexed: 01/12/2023]
Abstract
The identification of neurotransmitter type used by a neuron is important for the functional dissection of neuronal circuits. In the model organism Drosophila melanogaster, several methods for discerning the neurotransmitter systems are available. Here, we expanded the toolbox for the identification of cholinergic neurons by generating a new line FRT-STOP-FRT-VAChT::HA that is a conditional tagged knock-in of the vesicular acetylcholine transporter (VAChT) gene in its endogenous locus. Importantly, in comparison to already available tools for the detection of cholinergic neurons, the FRT-STOP-FRT-VAChT::HA allele also allows for identification of the subcellular localization of the cholinergic presynaptic release sites in a cell-specific manner. We used the newly generated FRT-STOP-FRT-VAChT::HA line to characterize the Mi1 and Tm3 neurons in the fly visual system and found that VAChT is present in the axons of both cell types, suggesting that Mi1 and Tm3 neurons provide cholinergic input to the elementary motion detectors, the T4 neurons. Summary: A new transgenic Drosophila melanogaster line for the cell-type-specific identification of cholinergic release sites expands the available methods toolbox for discerning the neurotransmitter systems in the fly nervous system.
Collapse
Affiliation(s)
- Katarina Pankova
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany .,Graduate School of Systemic Neurosciences, LMU Munich, 80539 Munich, Germany
| | - Alexander Borst
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| |
Collapse
|