1
|
Key J, Almaguer-Mederos LE, Kandi AR, Sen NE, Gispert S, Köpf G, Meierhofer D, Auburger G. ATXN2L primarily interacts with NUFIP2, the absence of ATXN2L results in NUFIP2 depletion, and the ATXN2-polyQ expansion triggers NUFIP2 accumulation. Neurobiol Dis 2025; 209:106903. [PMID: 40220918 DOI: 10.1016/j.nbd.2025.106903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
The cytoplasmic Ataxin-2 (ATXN2) protein associates with TDP-43 in stress granules (SG) where RNA quality control occurs. Mutations in this pathway underlie Spinocerebellar Ataxia type 2 (SCA2) and Amyotrophic Lateral Sclerosis. In contrast, Ataxin-2-like (ATXN2L) is predominantly perinuclear, more abundant, and essential for embryonic life. Its sequestration into ATXN2 aggregates may contribute to disease. In this study, we utilized two approaches to clarify the roles of ATXN2L. First, we identified interactors through co-immunoprecipitation in both wild-type and ATXN2L-null murine embryonic fibroblasts. Second, we assessed the proteome profile effects using mass spectrometry in these cells. Additionally, we examined the accumulation of ATXN2L interactors in the SCA2 mouse model, Atxn2-CAG100-KnockIn (KIN). We observed that RNA-binding proteins, including PABPN1, NUFIP2, MCRIP2, RBMS1, LARP1, PTBP1, FMR1, RPS20, FUBP3, MBNL2, ZMAT3, SFPQ, CSDE1, HNRNPK, and HNRNPDL, exhibit a stronger association with ATXN2L compared to established interactors like ATXN2, PABPC1, LSM12, and G3BP2. Additionally, ATXN2L interacted with components of the actin complex, such as SYNE2, LMOD1, ACTA2, FYB, and GOLGA3. We noted that oxidative stress increased HNRNPK but decreased SYNE2 association, which likely reflects the relocalization of SG. Proteome profiling revealed that NUFIP2 and SYNE2 are depleted in ATXN2L-null fibroblasts. Furthermore, NUFIP2 homodimers and SYNE1 accumulate during the ATXN2 aggregation process in KIN 14-month-old spinal cord tissues. The functions of ATXN2L and its interactors are therefore critical in RNA granule trafficking and surveillance, particularly for the maintenance of differentiated neurons.
Collapse
Affiliation(s)
- Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - Luis-Enrique Almaguer-Mederos
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - Arvind Reddy Kandi
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - Nesli-Ece Sen
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - Suzana Gispert
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - Gabriele Köpf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich- Hoffmann-Str. 7, 60528 Frankfurt am Main, Germany; Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Fachbereich Medizin, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
2
|
Taira Y, Zhu L, Fukunaga R. RNA-binding protein Miso/CG44249 is crucial for minor splicing during oogenesis in Drosophila. RNA (NEW YORK, N.Y.) 2025; 31:822-835. [PMID: 40169226 DOI: 10.1261/rna.080311.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/18/2025] [Indexed: 04/03/2025]
Abstract
Pre-mRNA introns are removed by two distinct spliceosomes: the major (U2-type) spliceosome, which splices over 99.5% of introns, and the minor (U12-type) spliceosome, responsible for a rare class of introns known as minor introns. While the major spliceosome contains U1, U2, U4, U5, and U6 small nuclear RNAs (snRNAs) along with numerous associated proteins, the minor spliceosome comprises U11, U12, U4atac, U5, and U6atac snRNAs and includes specialized proteins. The function and regulation of the minor spliceosome are critical. Mutations in its specific component, RNA-binding protein RNPC3/65K, are linked to human diseases such as primary ovarian insufficiency. In this study, we identify RNA-binding protein Miso (CG44249), which shares 31% and 27% amino acid sequence identity with human RNPC3 and RBM41, respectively, as a key factor in minor splicing and oogenesis in Drosophila Miso associates with U11 and U12 snRNAs in ovaries. miso mutant females exhibit smaller ovaries, reduced germline stem cell numbers, disrupted oogenesis, reduced fecundity, and lower fertility. In miso mutant ovaries, significant minor intron retention is observed, accompanied by a reduction in spliced RNAs and protein products. Our findings establish Miso as a critical factor for minor intron splicing and underscore its essential role in Drosophila oogenesis.
Collapse
Affiliation(s)
- Yuki Taira
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | - Li Zhu
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
3
|
Fiorenzani C, Mossa A, De Rubeis S. DEAD/DEAH-box RNA helicases shape the risk of neurodevelopmental disorders. Trends Genet 2025; 41:437-449. [PMID: 39828505 PMCID: PMC12055483 DOI: 10.1016/j.tig.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025]
Abstract
The DEAD/DEAH-box family of RNA helicases (RHs) is among the most abundant and conserved in eukaryotes. These proteins catalyze the remodeling of RNAs to regulate their splicing, stability, localization, and translation. Rare genetic variants in DEAD/DEAH-box proteins have recently emerged as being associated with neurodevelopmental disorders (NDDs). Analyses in cellular and animal models have uncovered fundamental roles for these proteins during brain development. We discuss the genetic and functional evidence that implicates DEAD/DEAH-box proteins in brain development and NDDs, with a focus on how structural insights from paralogous genes can be leveraged to advance our understanding of the pathogenic mechanisms at play.
Collapse
Affiliation(s)
- Chiara Fiorenzani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adele Mossa
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Zhang S, Zhang Y, Chen T, Hu HY, Lu C. The LSmAD Domain of Ataxin-2 Modulates the Structure and RNA Binding of Its Preceding LSm Domain. Cells 2025; 14:383. [PMID: 40072111 PMCID: PMC11898529 DOI: 10.3390/cells14050383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Ataxin-2 (Atx2), an RNA-binding protein, plays a pivotal role in the regulation of RNA, intracellular metabolism, and translation within the cellular environment. Although both the Sm-like (LSm) and LSm-associated (LSmAD) domains are considered to associated with RNA binding, there is still a lack of experimental evidence supporting their functions. To address this, we designed and constructed several recombinants containing the RNA-binding domain (RBD) of Atx2. By employing biophysical and biochemical techniques, such as EMSA and SHAPE chemical detection, we identified that LSm is responsible for RNA binding, whereas LSmAD alone does not bind RNA. NMR and small-angle X-ray scattering (SAXS) analyses have revealed that the LSmAD domain exhibits limited structural integrity and poor folding capability. The EMSA data confirmed that both LSm and LSm-LSmAD bind RNA, whereas LSmAD alone cannot, suggesting that LSmAD may serve as an auxiliary role to the LSm domain. SHAPE chemical probing further demonstrates that LSm binds to the AU-rich, GU-rich, or CU-rich sequence, but not to the CA-rich sequence. These findings indicate that Atx2 can interact with the U-rich sequences in the 3'-UTR, implicating its role in poly(A) tailing and the regulation of mRNA translation and degradation.
Collapse
Affiliation(s)
- Shengping Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; (S.Z.); (Y.Z.); (T.C.)
| | - Yunlong Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; (S.Z.); (Y.Z.); (T.C.)
| | - Ting Chen
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; (S.Z.); (Y.Z.); (T.C.)
| | - Hong-Yu Hu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Changrui Lu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; (S.Z.); (Y.Z.); (T.C.)
| |
Collapse
|
5
|
Teimuri S, Suter B. Drosophila Topoisomerase 3β binds to mRNAs in vivo, contributes to their localization and stability, and counteracts premature aging. PLoS One 2025; 20:e0318142. [PMID: 39932982 DOI: 10.1371/journal.pone.0318142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 01/12/2025] [Indexed: 02/13/2025] Open
Abstract
Topoisomerase 3β (Top3β) works not only on DNA but also on RNA. We isolated and identified the naturally cross-linked RNA targets of Drosophila Top3β from an early embryonic stage that contains almost exclusively maternal mRNAs. Favorite targets were long RNAs, particularly with long 3'UTRs, and RNAs that become localized in large cells. Top3β lacking only the hydroxyl group that makes the covalent bond to the RNA, did not allow normal expression and localization of Top3β mRNA targets or their protein products, demonstrating the importance of the enzymatic activity of Top3 β for optimized gene expression. Top3β is not essential for development to the adult stage but to maintain the morphology of the adult neuromuscular junction and to prevent premature loss of coordinated movement and aging. Alterations in human Top3β have been associated with several neurological diseases and cancers. The homologs of genes and (pre)mRNAs mis-expressed in these conditions show the same characteristics identified in the Drosophila Top3β targets, suggesting that Drosophila could model human Top3β. An in vivo test of this model showed that the enzymatic activity of Top3β reduces the neurodegeneration caused by the cytotoxic human (G4C2)49 RNA. Top3β supports normal gene expression, particularly of long and complex transcripts that must be transported and translationally controlled. These RNAs encode large cytoskeletal, cortical, and membrane proteins that are particularly important in large and long cells like motoneurons. Their reduced expression in the mutant seems to stress the cells, increasing the chances of developing neurodegenerative diseases.
Collapse
Affiliation(s)
- Shohreh Teimuri
- Institute of Cell Biology, University of Bern, Berne, Switzerland
| | - Beat Suter
- Institute of Cell Biology, University of Bern, Berne, Switzerland
| |
Collapse
|
6
|
Jeong YJ, Kim JC, Song G, Lee MR, Kim JS. RNA sequencing of Beauveria bassiana JEF-350-infected Thrips palmi reveals change of host defense and homeostasis. Appl Microbiol Biotechnol 2024; 108:514. [PMID: 39535617 PMCID: PMC11561002 DOI: 10.1007/s00253-024-13345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/14/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Melon thrips, Thrips palmi, represent a significant threat to plants, inducing necrosis and acting as vectors for numerous plant viruses. Entomopathogenic fungi present a promising avenue for the management of melon thrips populations resistant to conventional chemical treatments. In this work, an adult colony of melon thrips was exposed to Beauveria bassiana strain JEF-350, and the ensuing transcriptional response of the infected thrips was scrutinized to elucidate their reactions during fungal pathogenesis. Utilizing Illumina sequencing, RNA samples were extracted from untreated thrips as well as from thrips continuously infected for 2 and 4 days, each with three biological replicates. While no notable alterations in gene expression were observed between the untreated control and thrips infected for 2 days, those infected for 4 days exhibited a plethora of differentially expressed genes. Specifically, in the thrips infected for the extended period, pathways associated with lysosomal function and insect hormone biosynthesis were notably repressed, while others such as serine and glycine metabolism, Toll and Imd, and circadian rhythm pathways displayed heightened activity. Noteworthy downregulation was observed in numerous lysosomal hydrolase genes encoding glycosidases, sulfatases, and lipases, particularly glycosidases. Furthermore, certain genes related to hydrolase precursors within the Golgi apparatus exhibited heightened expression levels but failed to progress toward hydrolase biosynthesis. Upstream regulation of juvenile hormone biosynthesis was augmented, yet downstream genes were significantly downregulated, leading to a disruption in juvenile hormone production. Similarly, while cytochrome P450 genes in the downstream of ecdysone biosynthesis were upregulated, expressions of cholesterol desaturase and cytochrome P450 genes in the upstream were inhibited, consequently dampening ecdysone biosynthesis. The observed differential targeting of organs or pathways by B. bassiana JEF-350, in contrast to conventional chemicals primarily affecting neurotransmission and energy production, suggests its potential efficacy in managing resistant thrips populations. Consequently, integrating JEF-350 into the chemical spray regimen or incorporating it into tank-mix formulations with chemical insecticides emerges as a pragmatic approach within the realm of integrated pest management strategies. KEY POINTS: • Beauveria treatment inhibited lysosomal function and hormone synthesis in thrips. • Thrips serine/glycine metabolism, Toll and Imd, and circadian rhythm pathways were activated. • Upstream functions of thrips hormone biosynthesis increased, while downstream functions were suppressed. • Regarding biosynthesis of metabolites, this fungus targets other pathways with resistance management.
Collapse
Affiliation(s)
- Yu Jin Jeong
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Jong-Cheol Kim
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Gahyeon Song
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Mi Rong Lee
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Jae Su Kim
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Jeonbuk National University, Jeonju, South Korea.
| |
Collapse
|
7
|
Li L, Wang M, Huang L, Zheng X, Wang L, Miao H. Ataxin-2: a powerful RNA-binding protein. Discov Oncol 2024; 15:298. [PMID: 39039334 PMCID: PMC11263328 DOI: 10.1007/s12672-024-01158-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024] Open
Abstract
Ataxin-2 (ATXN2) was originally discovered in the context of spinocerebellar ataxia type 2 (SCA2), but it has become a key player in various neurodegenerative diseases. This review delves into the multifaceted roles of ATXN2 in human diseases, revealing its diverse molecular and cellular pathways. The impact of ATXN2 on diseases extends beyond functional outcomes; it mainly interacts with various RNA-binding proteins (RBPs) to regulate different stages of post-transcriptional gene expression in diseases. With the progress of research, ATXN2 has also been found to play an important role in the development of various cancers, including breast cancer, gastric cancer, pancreatic cancer, colon cancer, and esophageal cancer. This comprehensive exploration underscores the crucial role of ATXN2 in the pathogenesis of diseases and warrants further investigation by the scientific community. By reviewing the latest discoveries on the regulatory functions of ATXN2 in diseases, this article helps us understand the complex molecular mechanisms of a series of human diseases related to this intriguing protein.
Collapse
Affiliation(s)
- Lulu Li
- School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- Department of Clinical and Military Laboratory Medicine, College of Medical Laboratory Science, Army Medical University, Chongqing, 400038, China
| | - Meng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Lai Huang
- Department of Clinical and Military Laboratory Medicine, College of Medical Laboratory Science, Army Medical University, Chongqing, 400038, China
| | - Xiaoli Zheng
- School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Lina Wang
- Department of Clinical and Military Laboratory Medicine, College of Medical Laboratory Science, Army Medical University, Chongqing, 400038, China.
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
8
|
Costa RG, Conceição A, Matos CA, Nóbrega C. The polyglutamine protein ATXN2: from its molecular functions to its involvement in disease. Cell Death Dis 2024; 15:415. [PMID: 38877004 PMCID: PMC11178924 DOI: 10.1038/s41419-024-06812-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024]
Abstract
A CAG repeat sequence in the ATXN2 gene encodes a polyglutamine (polyQ) tract within the ataxin-2 (ATXN2) protein, showcasing a complex landscape of functions that have been progressively unveiled over recent decades. Despite significant progresses in the field, a comprehensive overview of the mechanisms governed by ATXN2 remains elusive. This multifaceted protein emerges as a key player in RNA metabolism, stress granules dynamics, endocytosis, calcium signaling, and the regulation of the circadian rhythm. The CAG overexpansion within the ATXN2 gene produces a protein with an extended poly(Q) tract, inducing consequential alterations in conformational dynamics which confer a toxic gain and/or partial loss of function. Although overexpanded ATXN2 is predominantly linked to spinocerebellar ataxia type 2 (SCA2), intermediate expansions are also implicated in amyotrophic lateral sclerosis (ALS) and parkinsonism. While the molecular intricacies await full elucidation, SCA2 presents ATXN2-associated pathological features, encompassing autophagy impairment, RNA-mediated toxicity, heightened oxidative stress, and disruption of calcium homeostasis. Presently, SCA2 remains incurable, with patients reliant on symptomatic and supportive treatments. In the pursuit of therapeutic solutions, various studies have explored avenues ranging from pharmacological drugs to advanced therapies, including cell or gene-based approaches. These endeavours aim to address the root causes or counteract distinct pathological features of SCA2. This review is intended to provide an updated compendium of ATXN2 functions, delineate the associated pathological mechanisms, and present current perspectives on the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Rafael G Costa
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal.
- PhD program in Biomedical Sciences, Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal.
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal.
| | - André Conceição
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
- PhD program in Biomedical Sciences, Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal
- Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal
- Champalimaud Research Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Carlos A Matos
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal.
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve (UAlg), Faro, Portugal.
| |
Collapse
|
9
|
Rangone H, Bond L, Weil TT, Glover DM. Greatwall-Endos-PP2A/B55 Twins network regulates translation and stability of maternal transcripts in the Drosophila oocyte-to-embryo transition. Open Biol 2024; 14:240065. [PMID: 38896085 DOI: 10.1098/rsob.240065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/24/2024] [Indexed: 06/21/2024] Open
Abstract
The transition from oocyte to embryo requires translation of maternally provided transcripts that in Drosophila is activated by Pan Gu kinase to release a rapid succession of 13 mitotic cycles. Mitotic entry is promoted by several protein kinases that include Greatwall/Mastl, whose Endosulfine substrates antagonize Protein Phosphatase 2A (PP2A), facilitating mitotic Cyclin-dependent kinase 1/Cyclin B kinase activity. Here we show that hyperactive greatwallScant can not only be suppressed by mutants in its Endos substrate but also by mutants in Pan Gu kinase subunits. Conversely, mutants in me31B or trailer hitch, which encode a complex that represses hundreds of maternal mRNAs, enhance greatwallScant . Me31B and Trailer Hitch proteins, known substrates of Pan Gu kinase, copurify with Endos. This echoes findings that budding yeast Dhh1, orthologue of Me31B, associates with Igo1/2, orthologues of Endos and substrates of the Rim15, orthologue of Greatwall. endos-derived mutant embryos show reduced Me31B and elevated transcripts for the mitotic activators Cyclin B, Polo and Twine/Cdc25. Together, our findings demonstrate a previously unappreciated conservation of the Greatwall-Endosulfine pathway in regulating translational repressors and its interactions with the Pan Gu kinase pathway to regulate translation and/or stability of maternal mRNAs upon egg activation.
Collapse
Affiliation(s)
- Hélène Rangone
- Department of Genetics, University of Cambridge, Downing Street , Cambridge, UK
| | - Laura Bond
- Department of Genetics, University of Cambridge, Downing Street , Cambridge, UK
| | - Timothy T Weil
- Department of Zoology, University of Cambridge, Downing Street , Cambridge, UK
| | - David M Glover
- Department of Genetics, University of Cambridge, Downing Street , Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Blvd , Pasadena, CA 91125, USA
| |
Collapse
|
10
|
Gao M. Me31B: a key repressor in germline regulation and beyond. Biosci Rep 2024; 44:BSR20231769. [PMID: 38606619 PMCID: PMC11065648 DOI: 10.1042/bsr20231769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/13/2024] Open
Abstract
Maternally Expressed at 31B (Me31B), an evolutionarily conserved ATP-dependent RNA helicase, plays an important role in the development of the germline across diverse animal species. Its cellular functionality has been posited as a translational repressor, participating in various RNA metabolism pathways to intricately regulate the spatiotemporal expression of RNAs. Despite its evident significance, the precise role and mechanistic underpinnings of Me31B remain insufficiently understood. This article endeavors to comprehensively review historic and recent research on Me31B, distill the major findings, discern generalizable patterns in Me31B's functions across different research contexts, and provide insights into its fundamental role and mechanism of action. The primary focus of this article centers on elucidating the role of Drosophila Me31B within the germline, while concurrently delving into pertinent research on its orthologs within other species and cellular systems.
Collapse
Affiliation(s)
- Ming Gao
- Biology Department, Indiana University Northwest, Gary, IN, U.S.A
| |
Collapse
|
11
|
Petrauskas A, Fortunati DL, Kandi AR, Pothapragada SS, Agrawal K, Singh A, Huelsmeier J, Hillebrand J, Brown G, Chaturvedi D, Lee J, Lim C, Auburger G, VijayRaghavan K, Ramaswami M, Bakthavachalu B. Structured and disordered regions of Ataxin-2 contribute differently to the specificity and efficiency of mRNP granule formation. PLoS Genet 2024; 20:e1011251. [PMID: 38768217 PMCID: PMC11166328 DOI: 10.1371/journal.pgen.1011251] [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: 01/27/2024] [Revised: 06/11/2024] [Accepted: 04/05/2024] [Indexed: 05/22/2024] Open
Abstract
Ataxin-2 (ATXN2) is a gene implicated in spinocerebellar ataxia type II (SCA2), amyotrophic lateral sclerosis (ALS) and Parkinsonism. The encoded protein is a therapeutic target for ALS and related conditions. ATXN2 (or Atx2 in insects) can function in translational activation, translational repression, mRNA stability and in the assembly of mRNP-granules, a process mediated by intrinsically disordered regions (IDRs). Previous work has shown that the LSm (Like-Sm) domain of Atx2, which can help stimulate mRNA translation, antagonizes mRNP-granule assembly. Here we advance these findings through a series of experiments on Drosophila and human Ataxin-2 proteins. Results of Targets of RNA Binding Proteins Identified by Editing (TRIBE), co-localization and immunoprecipitation experiments indicate that a polyA-binding protein (PABP) interacting, PAM2 motif of Ataxin-2 may be a major determinant of the mRNA and protein content of Ataxin-2 mRNP granules. Experiments with transgenic Drosophila indicate that while the Atx2-LSm domain may protect against neurodegeneration, structured PAM2- and unstructured IDR- interactions both support Atx2-induced cytotoxicity. Taken together, the data lead to a proposal for how Ataxin-2 interactions are remodelled during translational control and how structured and non-structured interactions contribute differently to the specificity and efficiency of RNP granule condensation as well as to neurodegeneration.
Collapse
Affiliation(s)
- Arnas Petrauskas
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - Daniel L. Fortunati
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - Arvind Reddy Kandi
- School of Biosciences and Bioengineering, Indian Institute of Technology, Mandi, India
| | | | - Khushboo Agrawal
- Tata Institute for Genetics and Society Centre at inStem, Bellary Road, Bangalore, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham University, Kollam, Kerala, India
| | - Amanjot Singh
- National Centre for Biological Sciences, TIFR, Bangalore, India
- Manipal Institute of Regenerative Medicine, MAHE-Bengaluru, Govindapura, Bengaluru, India
| | - Joern Huelsmeier
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - Jens Hillebrand
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - Georgia Brown
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | | | - Jongbo Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Ulsan, Republic of Korea
| | - Chunghun Lim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Ulsan, Republic of Korea
| | - Georg Auburger
- Experimental Neurology, Medical School, Goethe University, Frankfurt, Germany
| | | | - Mani Ramaswami
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
- National Centre for Biological Sciences, TIFR, Bangalore, India
| | - Baskar Bakthavachalu
- School of Biosciences and Bioengineering, Indian Institute of Technology, Mandi, India
- Tata Institute for Genetics and Society Centre at inStem, Bellary Road, Bangalore, India
| |
Collapse
|
12
|
Kara E, McCambridge A, Proffer M, Dilts C, Pumnea B, Eshak J, Smith KA, Fielder I, Doyle DA, Ortega BM, Mukatash Y, Malik N, Mohammed AR, Govani D, Niepielko MG, Gao M. Mutational analysis of the functional motifs of the DEAD-box RNA helicase Me31B/DDX6 in Drosophila germline development. FEBS Lett 2023; 597:1848-1867. [PMID: 37235728 PMCID: PMC10389067 DOI: 10.1002/1873-3468.14668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Me31B/DDX6 is a DEAD-box family RNA helicase playing roles in post-transcriptional RNA regulation in different cell types and species. Despite the known motifs/domains of Me31B, the in vivo functions of the motifs remain unclear. Here, we used the Drosophila germline as a model and used CRISPR to mutate the key Me31B motifs/domains: helicase domain, N-terminal domain, C-terminal domain and FDF-binding motif. Then, we performed screening characterization on the mutants and report the effects of the mutations on the Drosophila germline, on processes such as fertility, oogenesis, embryo patterning, germline mRNA regulation and Me31B protein expression. The study indicates that the Me31B motifs contribute different functions to the protein and are needed for proper germline development, providing insights into the in vivo working mechanism of the helicase.
Collapse
Affiliation(s)
- Evan Kara
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | | | - Megan Proffer
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Carol Dilts
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Brooke Pumnea
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - John Eshak
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Korey A. Smith
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Isaac Fielder
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Dominique A. Doyle
- School of Integrative Science and Technology, Kean University, Union, NJ, USA
| | - Bianca M. Ortega
- School of Integrative Science and Technology, Kean University, Union, NJ, USA
| | - Yousif Mukatash
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Noor Malik
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | | | - Deep Govani
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Matthew G. Niepielko
- School of Integrative Science and Technology, Kean University, Union, NJ, USA
- Biology Department, Kean University, Union, NJ, USA
| | - Ming Gao
- Biology Department, Indiana University Northwest, Gary, IN, USA
| |
Collapse
|
13
|
Steiner P, Arlt E, Boekhoff I, Gudermann T, Zierler S. TPC Functions in the Immune System. Handb Exp Pharmacol 2023; 278:71-92. [PMID: 36639434 DOI: 10.1007/164_2022_634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels (TPCs) are novel intracellular cation channels, which play a key role in numerous (patho-)physiological and immunological processes. In this chapter, we focus on their function in immune cells and immune reactions. Therefore, we first give an overview of the cellular immune response and the partaking immune cells. Second, we concentrate on ion channels which in the past have been shown to play an important role in the regulation of immune cells. The main focus is then directed to TPCs, which are primarily located in the membranes of acidic organelles, such as lysosomes or endolysosomes but also certain other vesicles. They regulate Ca2+ homeostasis and thus Ca2+ signaling in immune cells. Due to this important functional role, TPCs are enjoying increasing attention within the field of immunology in the last few decades but are also becoming more pertinent as pharmacological targets for the treatment of pro-inflammatory diseases such as allergic hypersensitivity. However, to uncover the precise molecular mechanism of TPCs in immune cell responses, further molecular, genetic, and ultrastructural investigations on TPCs are necessary, which then may pave the way to develop novel therapeutic strategies to treat diseases such as anaphylaxis more specifically.
Collapse
Affiliation(s)
- Philip Steiner
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Susanna Zierler
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria.
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
14
|
Nevoa JC, Latorre-Estivalis JM, Pais FSM, Marliére NP, Fernandes GDR, Lorenzo MG, Guarneri AA. Global characterization of gene expression in the brain of starved immature Rhodnius prolixus. PLoS One 2023; 18:e0282490. [PMID: 36867641 PMCID: PMC9983911 DOI: 10.1371/journal.pone.0282490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Rhodnius prolixus is a vector of Chagas disease and has become a model organism to study physiology, behavior, and pathogen interaction. The publication of its genome allowed initiating a process of comparative characterization of the gene expression profiles of diverse organs exposed to varying conditions. Brain processes control the expression of behavior and, as such, mediate immediate adjustment to a changing environment, allowing organisms to maximize their chances to survive and reproduce. The expression of fundamental behavioral processes like feeding requires fine control in triatomines because they obtain their blood meals from potential predators. Therefore, the characterization of gene expression profiles of key components modulating behavior in brain processes, like those of neuropeptide precursors and their receptors, seems fundamental. Here we study global gene expression profiles in the brain of starved R. prolixus fifth instar nymphs by means of RNA sequencing (RNA-Seq). RESULTS The expression of neuromodulatory genes such as those of precursors of neuropeptides, neurohormones, and their receptors; as well as the enzymes involved in the biosynthesis and processing of neuropeptides and biogenic amines were fully characterized. Other important gene targets such as neurotransmitter receptors, nuclear receptors, clock genes, sensory receptors, and takeouts genes were identified and their gene expression analyzed. CONCLUSION We propose that the set of neuromodulatory-related genes highly expressed in the brain of starved R. prolixus nymphs deserves functional characterization to allow the subsequent development of tools targeting them for bug control. As the brain is a complex structure that presents functionally specialized areas, future studies should focus on characterizing gene expression profiles in target areas, e.g. mushroom bodies, to complement our current knowledge.
Collapse
Affiliation(s)
- Jessica Coraiola Nevoa
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou – FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Jose Manuel Latorre-Estivalis
- Laboratorio de Insectos Sociales, Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires - CONICET, Buenos Aires, Argentina
| | | | - Newmar Pinto Marliére
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou – FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | | | - Marcelo Gustavo Lorenzo
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou – FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Alessandra Aparecida Guarneri
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou – FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
15
|
Patel S, Zissimopoulos S, Marchant JS. Endo-Lysosomal Two-Pore Channels and Their Protein Partners. Handb Exp Pharmacol 2023; 278:199-214. [PMID: 35902438 DOI: 10.1007/164_2022_601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels are ion channels expressed on acidic organelles such as the various vesicles that constitute the endo-lysosomal system. They are permeable to Ca2+ and Na+ and activated by the second messenger NAADP as well as the phosphoinositide, PI(3,5)P2 and/or voltage. Here, we review the proteins that interact with these channels including recently identified NAADP receptors.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK.
| | | | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
16
|
Nadimpalli HP, Guitart T, Coll O, Gebauer F. Ataxin-2, Twenty-four, and Dicer-2 are components of a noncanonical cytoplasmic polyadenylation complex. Life Sci Alliance 2022; 5:5/12/e202201417. [PMID: 36114004 PMCID: PMC9481931 DOI: 10.26508/lsa.202201417] [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: 02/18/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Identification of components of a noncanonical cytoplasmic polyadenylation machinery in Drosophila expands the diversity of RNA-binding proteins involved in poly(A) tail length control. Cytoplasmic polyadenylation is a mechanism to promote mRNA translation in a wide variety of biological contexts. A canonical complex centered around the conserved RNA-binding protein family CPEB has been shown to be responsible for this process. We have previously reported evidence for an alternative noncanonical, CPEB-independent complex in Drosophila, of which the RNA-interference factor Dicer-2 is a component. Here, we investigate Dicer-2 mRNA targets and protein cofactors in cytoplasmic polyadenylation. Using RIP-Seq analysis, we identify hundreds of potential Dicer-2 target transcripts, ∼60% of which were previously found as targets of the cytoplasmic poly(A) polymerase Wispy, suggesting widespread roles of Dicer-2 in cytoplasmic polyadenylation. Large-scale immunoprecipitation revealed Ataxin-2 and Twenty-four among the high-confidence interactors of Dicer-2. Complex analyses indicated that both factors form an RNA-independent complex with Dicer-2 and mediate interactions of Dicer-2 with Wispy. Functional poly(A)-test analyses showed that Twenty-four and Ataxin-2 are required for cytoplasmic polyadenylation of a subset of Dicer-2 targets. Our results reveal components of a novel cytoplasmic polyadenylation complex that operates during Drosophila early embryogenesis.
Collapse
Affiliation(s)
- Hima Priyanka Nadimpalli
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Tanit Guitart
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Olga Coll
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fátima Gebauer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- University of Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
17
|
Hart AF, Verbeeck J, Ariza D, Cejas D, Ghisbain G, Honchar H, Radchenko VG, Straka J, Ljubomirov T, Lecocq T, Dániel-Ferreira J, Flaminio S, Bortolotti L, Karise R, Meeus I, Smagghe G, Vereecken N, Vandamme P, Michez D, Maebe K. Signals of adaptation to agricultural stress in the genomes of two European bumblebees. Front Genet 2022; 13:993416. [PMID: 36276969 PMCID: PMC9579324 DOI: 10.3389/fgene.2022.993416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Human-induced environmental impacts on wildlife are widespread, causing major biodiversity losses. One major threat is agricultural intensification, typically characterised by large areas of monoculture, mechanical tillage, and the use of agrochemicals. Intensification leads to the fragmentation and loss of natural habitats, native vegetation, and nesting and breeding sites. Understanding the adaptability of insects to these changing environmental conditions is critical to predicting their survival. Bumblebees, key pollinators of wild and cultivated plants, are used as model species to assess insect adaptation to anthropogenic stressors. We investigated the effects of agricultural pressures on two common European bumblebees, Bombus pascuorum and B. lapidarius. Restriction-site Associated DNA Sequencing was used to identify loci under selective pressure across agricultural-natural gradients over 97 locations in Europe. 191 unique loci in B. pascuorum and 260 in B. lapidarius were identified as under selective pressure, and associated with agricultural stressors. Further investigation suggested several candidate proteins including several neurodevelopment, muscle, and detoxification proteins, but these have yet to be validated. These results provide insights into agriculture as a stressor for bumblebees, and signal for conservation action in light of ongoing anthropogenic changes.
Collapse
Affiliation(s)
- Alex F. Hart
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
| | - Jaro Verbeeck
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
| | - Daniel Ariza
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
| | - Diego Cejas
- Laboratory of Zoology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Guillaume Ghisbain
- Laboratory of Zoology, Research Institute for Biosciences, University of Mons, Mons, Belgium
- Smithsonian Tropical Research Institute, Gamboa, Panama
| | - Hanna Honchar
- Institute for Evolutionary Ecology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Vladimir G. Radchenko
- Institute for Evolutionary Ecology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Jakub Straka
- Charles University, Faculty of Science, Department of Zoology, Praha, Czech Republic
| | - Toshko Ljubomirov
- Institute of Biodiversity and Ecosystem Research—Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Thomas Lecocq
- Université de Lorraine, INRAE, URAFPA, Nancy, France
| | | | - Simone Flaminio
- Council for Agricultural Research and Economics, Research Centre for Agriculture and Environment, Bologna, Italy
| | - Laura Bortolotti
- Council for Agricultural Research and Economics, Research Centre for Agriculture and Environment, Bologna, Italy
| | - Reet Karise
- Estonian University of Life Sciences, Institute of Agricultural and Environmental Sciences, Tartu, Estonia
| | - Ivan Meeus
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
| | - Guy Smagghe
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
| | - Nicolas Vereecken
- Agroecology Lab, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Peter Vandamme
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Denis Michez
- Laboratory of Zoology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Kevin Maebe
- Ghent University, Faculty of Bioscience Engineering, Department of Plants and Crops, Lab of Agrozoology, Ghent, Belgium
- *Correspondence: Kevin Maebe,
| |
Collapse
|
18
|
Kim J, Muraoka M, Okada H, Toyoda A, Ajima R, Saga Y. The RNA helicase DDX6 controls early mouse embryogenesis by repressing aberrant inhibition of BMP signaling through miRNA-mediated gene silencing. PLoS Genet 2022; 18:e1009967. [PMID: 36197846 PMCID: PMC9534413 DOI: 10.1371/journal.pgen.1009967] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
The evolutionarily conserved RNA helicase DDX6 is a central player in post-transcriptional regulation, but its role during embryogenesis remains elusive. We here show that DDX6 enables proper cell lineage specification from pluripotent cells by analyzing Ddx6 knockout (KO) mouse embryos and employing an in vitro epiblast-like cell (EpiLC) induction system. Our study unveils that DDX6 is an important BMP signaling regulator. Deletion of Ddx6 causes the aberrant upregulation of the negative regulators of BMP signaling, which is accompanied by enhanced expression of Nodal and related genes. Ddx6 KO pluripotent cells acquire higher pluripotency with a strong inclination toward neural lineage commitment. During gastrulation, abnormally expanded Nodal and Eomes expression in the primitive streak likely promotes endoderm cell fate specification while inhibiting mesoderm differentiation. We also genetically dissected major DDX6 pathways by generating Dgcr8, Dcp2, and Eif4enif1 KO models in addition to Ddx6 KO. We found that the miRNA pathway mutant Dgcr8 KO phenocopies Ddx6 KO, indicating that DDX6 mostly works along with the miRNA pathway during early development, whereas its P-body-related functions are dispensable. Therefore, we conclude that DDX6 prevents aberrant upregulation of BMP signaling inhibitors by participating in miRNA-mediated gene silencing processes. Overall, this study delineates how DDX6 affects the development of the three primary germ layers during early mouse embryogenesis and the underlying mechanism of DDX6 function.
Collapse
Affiliation(s)
- Jessica Kim
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Masafumi Muraoka
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
| | - Hajime Okada
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Mishima, Japan
| | - Rieko Ajima
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
- Department of Genetics, The Graduate University for Advanced Studies, SOKENDAI, Mishima, Japan
- * E-mail: (RA); (YS)
| | - Yumiko Saga
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
- Department of Genetics, The Graduate University for Advanced Studies, SOKENDAI, Mishima, Japan
- * E-mail: (RA); (YS)
| |
Collapse
|
19
|
Zhang L, Zhang S, Wang R, Sun L. Genome-Wide Identification of Long Noncoding RNA and Their Potential Interactors in ISWI Mutants. Int J Mol Sci 2022; 23:ijms23116247. [PMID: 35682924 PMCID: PMC9181106 DOI: 10.3390/ijms23116247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been identified as key regulators of gene expression and participate in many vital physiological processes. Chromatin remodeling, being an important epigenetic modification, has been identified in many biological activities as well. However, the regulatory mechanism of lncRNA in chromatin remodeling remains unclear. In order to characterize the genome-wide lncRNA expression and their potential interacting factors during this process in Drosophila, we investigated the expression pattern of lncRNAs and mRNAs based on the transcriptome analyses and found significant differences between lncRNAs and mRNAs. Then, we performed TSA-FISH experiments of candidate lncRNAs and their potential interactors that have different functions in Drosophila embryos to determine their expression pattern. In addition, we also analyzed the expression of transposable elements (TEs) and their interactors to explore their expression in ISWI mutants. Our results provide a new perspective for understanding the possible regulatory mechanism of lncRNAs and TEs as well as their targets in chromatin remodeling.
Collapse
|
20
|
Abstract
The 5'-terminal cap is a fundamental determinant of eukaryotic gene expression which facilitates cap-dependent translation and protects mRNAs from exonucleolytic degradation. Enzyme-directed hydrolysis of the cap (decapping) decisively affects mRNA expression and turnover, and is a heavily regulated event. Following the identification of the decapping holoenzyme (Dcp1/2) over two decades ago, numerous studies revealed the complexity of decapping regulation across species and cell types. A conserved set of Dcp1/2-associated proteins, implicated in decapping activation and molecular scaffolding, were identified through genetic and molecular interaction studies, and yet their exact mechanisms of action are only emerging. In this review, we discuss the prevailing models on the roles and assembly of decapping co-factors, with considerations of conservation across species and comparison across physiological contexts. We next discuss the functional convergences of decapping machineries with other RNA-protein complexes in cytoplasmic P bodies and compare current views on their impact on mRNA stability and translation. Lastly, we review the current models of decapping activation and highlight important gaps in our current understanding.
Collapse
Affiliation(s)
- Elva Vidya
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Thomas F. Duchaine
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| |
Collapse
|
21
|
Yildirim E, Curtis R, Hwangbo DS. Roles of peripheral clocks: lessons from the fly. FEBS Lett 2022; 596:263-293. [PMID: 34862983 PMCID: PMC8844272 DOI: 10.1002/1873-3468.14251] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
To adapt to and anticipate rhythmic changes in the environment such as daily light-dark and temperature cycles, internal timekeeping mechanisms called biological clocks evolved in a diverse set of organisms, from unicellular bacteria to humans. These biological clocks play critical roles in organisms' fitness and survival by temporally aligning physiological and behavioral processes to the external cues. The central clock is located in a small subset of neurons in the brain and drives daily activity rhythms, whereas most peripheral tissues harbor their own clock systems, which generate metabolic and physiological rhythms. Since the discovery of Drosophila melanogaster clock mutants in the early 1970s, the fruit fly has become an extensively studied model organism to investigate the mechanism and functions of circadian clocks. In this review, we primarily focus on D. melanogaster to survey key discoveries and progresses made over the past two decades in our understanding of peripheral clocks. We discuss physiological roles and molecular mechanisms of peripheral clocks in several different peripheral tissues of the fly.
Collapse
Affiliation(s)
| | - Rachel Curtis
- Department of Biology, University of Louisville, Louisville, KY, USA
| | - Dae-Sung Hwangbo
- Department of Biology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
22
|
Rounds JC, Corgiat EB, Ye C, Behnke JA, Kelly SM, Corbett AH, Moberg KH. The disease-associated proteins Drosophila Nab2 and Ataxin-2 interact with shared RNAs and coregulate neuronal morphology. Genetics 2022; 220:iyab175. [PMID: 34791182 PMCID: PMC8733473 DOI: 10.1093/genetics/iyab175] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/27/2021] [Indexed: 01/05/2023] Open
Abstract
Nab2 encodes the Drosophila melanogaster member of a conserved family of zinc finger polyadenosine RNA-binding proteins (RBPs) linked to multiple steps in post-transcriptional regulation. Mutation of the Nab2 human ortholog ZC3H14 gives rise to an autosomal recessive intellectual disability but understanding of Nab2/ZC3H14 function in metazoan nervous systems is limited, in part because no comprehensive identification of metazoan Nab2/ZC3H14-associated RNA transcripts has yet been conducted. Moreover, many Nab2/ZC3H14 functional protein partnerships remain unidentified. Here, we present evidence that Nab2 genetically interacts with Ataxin-2 (Atx2), which encodes a neuronal translational regulator, and that these factors coordinately regulate neuronal morphology, circadian behavior, and adult viability. We then present the first high-throughput identifications of Nab2- and Atx2-associated RNAs in Drosophila brain neurons using RNA immunoprecipitation-sequencing (RIP-Seq). Critically, the RNA interactomes of each RBP overlap, and Nab2 exhibits high specificity in its RNA associations in neurons in vivo, associating with a small fraction of all polyadenylated RNAs. The identities of shared associated transcripts (e.g., drk, me31B, stai) and of transcripts specific to Nab2 or Atx2 (e.g., Arpc2 and tea) promise insight into neuronal functions of, and genetic interactions between, each RBP. Consistent with prior biochemical studies, Nab2-associated neuronal RNAs are overrepresented for internal A-rich motifs, suggesting these sequences may partially mediate Nab2 target selection. These data support a model where Nab2 functionally opposes Atx2 in neurons, demonstrate Nab2 shares associated neuronal RNAs with Atx2, and reveal Drosophila Nab2 associates with a more specific subset of polyadenylated mRNAs than its polyadenosine affinity alone may suggest.
Collapse
Affiliation(s)
- J Christopher Rounds
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Edwin B Corgiat
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Changtian Ye
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joseph A Behnke
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Seth M Kelly
- Department of Biology, The College of Wooster, Wooster, OH 44691, USA
| | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Kenneth H Moberg
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
23
|
Jeong J, Lee J, Kim JH, Lim C. Metabolic flux from the Krebs cycle to glutamate transmission tunes a neural brake on seizure onset. PLoS Genet 2021; 17:e1009871. [PMID: 34714823 PMCID: PMC8555787 DOI: 10.1371/journal.pgen.1009871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/11/2021] [Indexed: 01/18/2023] Open
Abstract
Kohlschütter-Tönz syndrome (KTS) manifests as neurological dysfunctions, including early-onset seizures. Mutations in the citrate transporter SLC13A5 are associated with KTS, yet their underlying mechanisms remain elusive. Here, we report that a Drosophila SLC13A5 homolog, I'm not dead yet (Indy), constitutes a neurometabolic pathway that suppresses seizure. Loss of Indy function in glutamatergic neurons caused "bang-induced" seizure-like behaviors. In fact, glutamate biosynthesis from the citric acid cycle was limiting in Indy mutants for seizure-suppressing glutamate transmission. Oral administration of the rate-limiting α-ketoglutarate in the metabolic pathway rescued low glutamate levels in Indy mutants and ameliorated their seizure-like behaviors. This metabolic control of the seizure susceptibility was mapped to a pair of glutamatergic neurons, reversible by optogenetic controls of their activity, and further relayed onto fan-shaped body neurons via the ionotropic glutamate receptors. Accordingly, our findings reveal a micro-circuit that links neural metabolism to seizure, providing important clues to KTS-associated neurodevelopmental deficits.
Collapse
Affiliation(s)
- Jiwon Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jongbin Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ji-hyung Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chunghun Lim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|
24
|
McIntosh CS, Li D, Wilton SD, Aung-Htut MT. Polyglutamine Ataxias: Our Current Molecular Understanding and What the Future Holds for Antisense Therapies. Biomedicines 2021; 9:1499. [PMID: 34829728 PMCID: PMC8615177 DOI: 10.3390/biomedicines9111499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Polyglutamine (polyQ) ataxias are a heterogenous group of neurological disorders all caused by an expanded CAG trinucleotide repeat located in the coding region of each unique causative gene. To date, polyQ ataxias encompass six disorders: spinocerebellar ataxia types 1, 2, 3, 6, 7, and 17 and account for a larger group of disorders simply known as polyglutamine disorders, which also includes Huntington's disease. These diseases are typically characterised by progressive ataxia, speech and swallowing difficulties, lack of coordination and gait, and are unfortunately fatal in nature, with the exception of SCA6. All the polyQ spinocerebellar ataxias have a hallmark feature of neuronal aggregations and share many common pathogenic mechanisms, such as mitochondrial dysfunction, impaired proteasomal function, and autophagy impairment. Currently, therapeutic options are limited, with no available treatments that slow or halt disease progression. Here, we discuss the common molecular and clinical presentations of polyQ spinocerebellar ataxias. We will also discuss the promising antisense oligonucleotide therapeutics being developed as treatments for these devastating diseases. With recent advancements and therapeutic approvals of various antisense therapies, it is envisioned that some of the studies reviewed may progress into clinical trials and beyond.
Collapse
Affiliation(s)
- Craig S. McIntosh
- Molecular Therapy Laboratory, Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute Murdoch University, Discovery Way, Murdoch, WA 6150, Australia; (C.S.M.); (D.L.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Dunhui Li
- Molecular Therapy Laboratory, Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute Murdoch University, Discovery Way, Murdoch, WA 6150, Australia; (C.S.M.); (D.L.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Steve D. Wilton
- Molecular Therapy Laboratory, Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute Murdoch University, Discovery Way, Murdoch, WA 6150, Australia; (C.S.M.); (D.L.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| | - May T. Aung-Htut
- Molecular Therapy Laboratory, Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute Murdoch University, Discovery Way, Murdoch, WA 6150, Australia; (C.S.M.); (D.L.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW To provide an update on the role of Ataxin-2 gene (ATXN2) in health and neurological diseases. RECENT FINDINGS There is a growing complexity emerging on the role of ATXN2 and its variants in association with SCA2 and several other neurological diseases. Polymorphisms and intermediate alleles in ATXN2 establish this gene as a powerful modulator of neurological diseases including lethal neurodegenerative conditions such as motor neuron disease, spinocerebellar ataxia 3 (SCA3), and peripheral nerve disease such as familial amyloidosis polyneuropathy. This role is in fact far wider than the previously described for polymorphism in the prion protein (PRNP) gene. Positive data from antisense oligo therapy in a murine model of SCA2 suggest that similar approaches may be feasible in humans SCA2 patients. SUMMARY ATXN2 is one of the few genes where a single gene causes several diseases and/or modifies several and disparate neurological disorders. Hence, understanding mutagenesis, genetic variants, and biological functions will help managing SCA2, and several human diseases connected with dysfunctional pathways in the brain, innate immunity, autophagy, cellular, lipid, and RNA metabolism.
Collapse
Affiliation(s)
- Jose Miguel Laffita-Mesa
- Department of Clinical Neuroscience (CNS), J5:20 Bioclinicum, Karolinska University Hospital, Stockholm, Sweden
| | | | | |
Collapse
|
26
|
Formicola N, Heim M, Dufourt J, Lancelot AS, Nakamura A, Lagha M, Besse F. Tyramine induces dynamic RNP granule remodeling and translation activation in the Drosophila brain. eLife 2021; 10:65742. [PMID: 33890854 PMCID: PMC8064753 DOI: 10.7554/elife.65742] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/04/2021] [Indexed: 02/06/2023] Open
Abstract
Ribonucleoprotein (RNP) granules are dynamic condensates enriched in regulatory RNA binding proteins (RBPs) and RNAs under tight spatiotemporal control. Extensive recent work has investigated the molecular principles underlying RNP granule assembly, unraveling that they form through the self-association of RNP components into dynamic networks of interactions. How endogenous RNP granules respond to external stimuli to regulate RNA fate is still largely unknown. Here, we demonstrate through high-resolution imaging of intact Drosophila brains that Tyramine induces a reversible remodeling of somatic RNP granules characterized by the decondensation of granule-enriched RBPs (e.g. Imp/ZBP1/IGF2BP) and helicases (e.g. Me31B/DDX-6/Rck). Furthermore, our functional analysis reveals that Tyramine signals both through its receptor TyrR and through the calcium-activated kinase CamkII to trigger RNP component decondensation. Finally, we uncover that RNP granule remodeling is accompanied by the rapid and specific translational activation of associated mRNAs. Thus, this work sheds new light on the mechanisms controlling cue-induced rearrangement of physiological RNP condensates.
Collapse
Affiliation(s)
- Nadia Formicola
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Marjorie Heim
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Jérémy Dufourt
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Montpellier, France
| | - Anne-Sophie Lancelot
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Akira Nakamura
- Department of Germline Development, Institute of Molecular Embryology and Genetics, and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mounia Lagha
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Montpellier, France
| | - Florence Besse
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| |
Collapse
|
27
|
The ATXN2 Orthologs CID3 and CID4, Act Redundantly to In-Fluence Developmental Pathways throughout the Life Cycle of Arabidopsis thaliana. Int J Mol Sci 2021; 22:ijms22063068. [PMID: 33802796 PMCID: PMC8002431 DOI: 10.3390/ijms22063068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022] Open
Abstract
RNA-binding proteins (RBPs) are key elements involved in post-transcriptional regulation. Ataxin-2 (ATXN2) is an evolutionarily conserved RBP protein, whose function has been studied in several model organisms, from Saccharomyces cerevisiae to the Homo sapiens. ATXN2 interacts with poly(A) binding proteins (PABP) and binds to specific sequences at the 3'UTR of target mRNAs to stabilize them. CTC-Interacting Domain3 (CID3) and CID4 are two ATXN2 orthologs present in plant genomes whose function is unknown. In the present study, phenotypical and transcriptome profiling were used to examine the role of CID3 and CID4 in Arabidopsis thaliana. We found that they act redundantly to influence pathways throughout the life cycle. cid3cid4 double mutant showed a delay in flowering time and a reduced rosette size. Transcriptome profiling revealed that key factors that promote floral transition and floral meristem identity were downregulated in cid3cid4 whereas the flowering repressor FLOWERING LOCUS C (FLC) was upregulated. Expression of key factors in the photoperiodic regulation of flowering and circadian clock pathways, were also altered in cid3cid4, as well as the expression of several transcription factors and miRNAs encoding genes involved in leaf growth dynamics. These findings reveal that ATXN2 orthologs may have a role in developmental pathways throughout the life cycle of plants.
Collapse
|
28
|
Singh A, Hulsmeier J, Kandi AR, Pothapragada SS, Hillebrand J, Petrauskas A, Agrawal K, RT K, Thiagarajan D, Jayaprakashappa D, VijayRaghavan K, Ramaswami M, Bakthavachalu B. Antagonistic roles for Ataxin-2 structured and disordered domains in RNP condensation. eLife 2021; 10:e60326. [PMID: 33689682 PMCID: PMC7946432 DOI: 10.7554/elife.60326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Ataxin-2 (Atx2) is a translational control molecule mutated in spinocerebellar ataxia type II and amyotrophic lateral sclerosis. While intrinsically disordered domains (IDRs) of Atx2 facilitate mRNP condensation into granules, how IDRs work with structured domains to enable positive and negative regulation of target mRNAs remains unclear. Using the Targets of RNA-Binding Proteins Identified by Editing technology, we identified an extensive data set of Atx2-target mRNAs in the Drosophila brain and S2 cells. Atx2 interactions with AU-rich elements in 3'UTRs appear to modulate stability/turnover of a large fraction of these target mRNAs. Further genomic and cell biological analyses of Atx2 domain deletions demonstrate that Atx2 (1) interacts closely with target mRNAs within mRNP granules, (2) contains distinct protein domains that drive or oppose RNP-granule assembly, and (3) has additional essential roles outside of mRNP granules. These findings increase the understanding of neuronal translational control mechanisms and inform strategies for Atx2-based interventions under development for neurodegenerative disease.
Collapse
Affiliation(s)
- Amanjot Singh
- National Centre for Biological SciencesBangaloreIndia
| | - Joern Hulsmeier
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College DublinDublinIreland
| | - Arvind Reddy Kandi
- National Centre for Biological SciencesBangaloreIndia
- Tata Institute for Genetics and Society Centre at inStem, Bellary RoadBangaloreIndia
| | | | - Jens Hillebrand
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College DublinDublinIreland
| | - Arnas Petrauskas
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College DublinDublinIreland
| | - Khushboo Agrawal
- Tata Institute for Genetics and Society Centre at inStem, Bellary RoadBangaloreIndia
- School of Biotechnology, Amrita Vishwa Vidyapeetham UniversityKollamIndia
| | - Krishnan RT
- National Centre for Biological SciencesBangaloreIndia
| | | | | | | | - Mani Ramaswami
- National Centre for Biological SciencesBangaloreIndia
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College DublinDublinIreland
| | - Baskar Bakthavachalu
- National Centre for Biological SciencesBangaloreIndia
- Tata Institute for Genetics and Society Centre at inStem, Bellary RoadBangaloreIndia
- School of Basic Sciences, Indian Institute of TechnologyMandiIndia
| |
Collapse
|
29
|
Lee J, Park J, Kim JH, Lee G, Park TE, Yoon KJ, Kim YK, Lim C. LSM12-EPAC1 defines a neuroprotective pathway that sustains the nucleocytoplasmic RAN gradient. PLoS Biol 2020; 18:e3001002. [PMID: 33362237 PMCID: PMC7757817 DOI: 10.1371/journal.pbio.3001002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nucleocytoplasmic transport (NCT) defects have been implicated in neurodegenerative diseases such as C9ORF72-associated amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we identify a neuroprotective pathway of like-Sm protein 12 (LSM12) and exchange protein directly activated by cyclic AMP 1 (EPAC1) that sustains the nucleocytoplasmic RAN gradient and thereby suppresses NCT dysfunction by the C9ORF72-derived poly(glycine-arginine) protein. LSM12 depletion in human neuroblastoma cells aggravated poly(GR)-induced impairment of NCT and nuclear integrity while promoting the nuclear accumulation of poly(GR) granules. In fact, LSM12 posttranscriptionally up-regulated EPAC1 expression, whereas EPAC1 overexpression rescued the RAN gradient and NCT defects in LSM12-deleted cells. C9-ALS patient-derived neurons differentiated from induced pluripotent stem cells (C9-ALS iPSNs) displayed low expression of LSM12 and EPAC1. Lentiviral overexpression of LSM12 or EPAC1 indeed restored the RAN gradient, mitigated the pathogenic mislocalization of TDP-43, and suppressed caspase-3 activation for apoptosis in C9-ALS iPSNs. EPAC1 depletion biochemically dissociated RAN-importin β1 from the cytoplasmic nuclear pore complex, thereby dissipating the nucleocytoplasmic RAN gradient essential for NCT. These findings define the LSM12-EPAC1 pathway as an important suppressor of the NCT-related pathologies in C9-ALS/FTD. A post-transcriptional circuit comprising LSM12 and EPAC1 suppresses neurodegenerative pathologies in C9ORF72-associated amyotrophic lateral sclerosis by establishing the RAN gradient and sustaining nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Jongbo Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jumin Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ji-hyung Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Giwook Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Tae-Eun Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yoon Ki Kim
- Creative Research Initiatives Center for Molecular Biology of Translation, Korea University, Seoul, Republic of Korea
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|
30
|
Key J, Harter PN, Sen NE, Gradhand E, Auburger G, Gispert S. Mid-Gestation lethality of Atxn2l-Ablated Mice. Int J Mol Sci 2020; 21:E5124. [PMID: 32698485 PMCID: PMC7404131 DOI: 10.3390/ijms21145124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Depletion of yeast/fly Ataxin-2 rescues TDP-43 overexpression toxicity. In mouse models of Amyotrophic Lateral Sclerosis via TDP-43 overexpression, depletion of its ortholog ATXN2 mitigated motor neuron degeneration and extended lifespan from 25 days to >300 days. There is another ortholog in mammals, named ATXN2L (Ataxin-2-like), which is almost uncharacterized but also functions in RNA surveillance at stress granules. We generated mice with Crispr/Cas9-mediated deletion of Atxn2l exons 5-8, studying homozygotes prenatally and heterozygotes during aging. Our novel findings indicate that ATXN2L absence triggers mid-gestational embryonic lethality, affecting female animals more strongly. Weight and development stages of homozygous mutants were reduced. Placenta phenotypes were not apparent, but brain histology showed lamination defects and apoptosis. Aged heterozygotes showed no locomotor deficits or weight loss over 12 months. Null mutants in vivo displayed compensatory efforts to maximize Atxn2l expression, which were prevented upon nutrient abundance in vitro. Mouse embryonal fibroblast cells revealed more multinucleated giant cells upon ATXN2L deficiency. In addition, in human neural cells, transcript levels of ATXN2L were induced upon starvation and glucose and amino acids exposure, but this induction was partially prevented by serum or low cholesterol administration. Neither ATXN2L depletion triggered dysregulation of ATXN2, nor a converse effect was observed. Overall, this essential role of ATXN2L for embryogenesis raises questions about its role in neurodegenerative diseases and neuroprotective therapies.
Collapse
Affiliation(s)
- Jana Key
- Exp. Neurology, Medical Faculty, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (J.K.); (N.-E.S.)
- Faculty of Biosciences, Goethe-University, Altenhöferallee 1, 60438 Frankfurt am Main, Germany
| | - Patrick N. Harter
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, Heinrich-Hoffmann-Strasse 7, 60528 Frankfurt am Main, Germany;
| | - Nesli-Ece Sen
- Exp. Neurology, Medical Faculty, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (J.K.); (N.-E.S.)
- Faculty of Biosciences, Goethe-University, Altenhöferallee 1, 60438 Frankfurt am Main, Germany
| | - Elise Gradhand
- Dr. Senckenberg Institute for Pathology, University Hospital, Goethe University, Theodor-Stern-Kai-7, 60590 Frankfurt am Main, Germany;
| | - Georg Auburger
- Exp. Neurology, Medical Faculty, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (J.K.); (N.-E.S.)
| | - Suzana Gispert
- Exp. Neurology, Medical Faculty, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (J.K.); (N.-E.S.)
| |
Collapse
|
31
|
Melo do Nascimento L, Terrao M, Marucha KK, Liu B, Egler F, Clayton C. The RNA-associated proteins MKT1 and MKT1L form alternative PBP1-containing complexes in Trypanosoma brucei. J Biol Chem 2020; 295:10940-10955. [PMID: 32532821 DOI: 10.1074/jbc.ra120.013306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/03/2020] [Indexed: 01/20/2023] Open
Abstract
Control of gene expression in kinetoplastids such as trypanosomes depends heavily on RNA-binding proteins that influence mRNA decay and translation. We previously showed that the trypanosome protein MKT1 forms a multicomponent protein complex: MKT1 interacts with PBP1, which in turn recruits LSM12 and poly(A)-binding protein. MKT1 is recruited to mRNAs by sequence-specific RNA-binding proteins, resulting in stabilization of the bound mRNA. We here show that PBP1, LSM12, and a 117-residue protein, XAC1 (Tb927.7.2780), are present in complexes that contain either MKT1 or an MKT1-like protein, MKT1L (Tb927.10.1490). All five proteins are present predominantly in the complexes, and we found evidence for a minor subset of complexes containing both MKT1 and MKT1L. XAC1-containing complexes reproducibly contained RNA-binding proteins that were previously found associated with MKT1. Moreover, XAC1- or MKT1-containing complexes specifically recruited one of the two poly(A)-binding proteins, PABP2, and one of the six cap-binding translation initiation complexes, EIF4E6-EIF4G5. Yeast two-hybrid assay results indicated that MKT1 directly interacts with EIF4G5. MKT1-PBP1 complexes can therefore interact with mRNAs via their poly(A) tails and caps, as well as through sequence-specific RNA-binding proteins. Correspondingly, MKT1 is associated with many mRNAs, although not with those encoding ribosomal proteins. Meanwhile, MKT1L resembles MKT1 at the C terminus but additionally features an N-terminal extension with low-complexity regions. Although MKT1L depletion inhibited cell proliferation, we found no evidence that it specifically interacts with RNA-binding proteins or mRNA. We speculate that MKT1L may compete with MKT1 for PBP1 binding and thereby modulate the function of MKT1-containing complexes.
Collapse
Affiliation(s)
| | - Monica Terrao
- Heidelberg University Centre for Molecular Biology (ZMBH), Heidelberg, Germany
| | | | - Bin Liu
- Heidelberg University Centre for Molecular Biology (ZMBH), Heidelberg, Germany
| | - Franziska Egler
- Heidelberg University Centre for Molecular Biology (ZMBH), Heidelberg, Germany
| | - Christine Clayton
- Heidelberg University Centre for Molecular Biology (ZMBH), Heidelberg, Germany
| |
Collapse
|
32
|
Taglini F, Chapman E, van Nues R, Theron E, Bayne EH. Mkt1 is required for RNAi-mediated silencing and establishment of heterochromatin in fission yeast. Nucleic Acids Res 2020; 48:1239-1253. [PMID: 31822915 PMCID: PMC7026591 DOI: 10.1093/nar/gkz1157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/12/2019] [Accepted: 11/28/2019] [Indexed: 01/04/2023] Open
Abstract
Constitutive domains of repressive heterochromatin are maintained within the fission yeast genome through self-reinforcing mechanisms involving histone methylation and small RNAs. Non-coding RNAs generated from heterochromatic regions are processed into small RNAs by the RNA interference pathway, and are subject to silencing through both transcriptional and post-transcriptional mechanisms. While the pathways involved in maintenance of the repressive heterochromatin state are reasonably well understood, less is known about the requirements for its establishment. Here, we describe a novel role for the post-transcriptional regulatory factor Mkt1 in establishment of heterochromatin at pericentromeres in fission yeast. Loss of Mkt1 does not affect maintenance of existing heterochromatin, but does affect its recovery following depletion, as well as de novo establishment of heterochromatin on a mini-chromosome. Pathway dissection revealed that Mkt1 is required for RNAi-mediated post-transcriptional silencing, downstream of small RNA production. Mkt1 physically associates with pericentromeric transcripts, and is additionally required for maintenance of silencing and heterochromatin at centromeres when transcriptional silencing is impaired. Our findings provide new insight into the mechanism of RNAi-mediated post-transcriptional silencing in fission yeast, and unveil an important role for post-transcriptional silencing in establishment of heterochromatin that is dispensable when full transcriptional silencing is imposed.
Collapse
Affiliation(s)
- Francesca Taglini
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Elliott Chapman
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Rob van Nues
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Emmanuelle Theron
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Elizabeth H Bayne
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
33
|
Abstract
Circadian clocks drive daily rhythms of physiology and behavior in multiple organisms and synchronize these rhythms to environmental cycles of light and temperature. The basic mechanism of the clock consists of a transcription-translation feedback loop, in which key clock proteins negatively regulate their own transcription. Although much of the focus with respect to clock mechanisms has been on the regulation of transcription and on the stability and activity of clock proteins, it is clear that other regulatory processes also have to be involved to explain aspects of clock function. Here, we review the role of alternative splicing in circadian clocks. Starting with a discussion of the Drosophila clock and then extending to other major circadian model systems, we describe how the control of alternative splicing enables organisms to maintain their circadian clocks as well as to respond to environmental inputs, in particular to temperature changes.
Collapse
Affiliation(s)
- Iryna Shakhmantsir
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amita Sehgal
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Foley LE, Ling J, Joshi R, Evantal N, Kadener S, Emery P. Drosophila PSI controls circadian period and the phase of circadian behavior under temperature cycle via tim splicing. eLife 2019; 8:50063. [PMID: 31702555 PMCID: PMC6890465 DOI: 10.7554/elife.50063] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/07/2019] [Indexed: 12/30/2022] Open
Abstract
The Drosophila circadian pacemaker consists of transcriptional feedback loops subjected to post-transcriptional and post-translational regulation. While post-translational regulatory mechanisms have been studied in detail, much less is known about circadian post-transcriptional control. Thus, we targeted 364 RNA binding and RNA associated proteins with RNA interference. Among the 43 hits we identified was the alternative splicing regulator P-element somatic inhibitor (PSI). PSI regulates the thermosensitive alternative splicing of timeless (tim), promoting splicing events favored at warm temperature over those increased at cold temperature. Psi downregulation shortens the period of circadian rhythms and advances the phase of circadian behavior under temperature cycle. Interestingly, both phenotypes were suppressed in flies that could produce TIM proteins only from a transgene that cannot form the thermosensitive splicing isoforms. Therefore, we conclude that PSI regulates the period of Drosophila circadian rhythms and circadian behavior phase during temperature cycling through its modulation of the tim splicing pattern.
Collapse
Affiliation(s)
- Lauren E Foley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Jinli Ling
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Radhika Joshi
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | | | - Sebastian Kadener
- Hebrew University of Jerusalem, Jerusalem, Israel.,Brandeis University, Waltham, United States
| | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
35
|
Xu F, Kula-Eversole E, Iwanaszko M, Lim C, Allada R. Ataxin2 functions via CrebA to mediate Huntingtin toxicity in circadian clock neurons. PLoS Genet 2019; 15:e1008356. [PMID: 31593562 PMCID: PMC6782096 DOI: 10.1371/journal.pgen.1008356] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Disrupted circadian rhythms is a prominent and early feature of neurodegenerative diseases including Huntington’s disease (HD). In HD patients and animal models, striatal and hypothalamic neurons expressing molecular circadian clocks are targets of mutant Huntingtin (mHtt) pathogenicity. Yet how mHtt disrupts circadian rhythms remains unclear. In a genetic screen for modifiers of mHtt effects on circadian behavior in Drosophila, we discovered a role for the neurodegenerative disease gene Ataxin2 (Atx2). Genetic manipulations of Atx2 modify the impact of mHtt on circadian behavior as well as mHtt aggregation and demonstrate a role for Atx2 in promoting mHtt aggregation as well as mHtt-mediated neuronal dysfunction. RNAi knockdown of the Fragile X mental retardation gene, dfmr1, an Atx2 partner, also partially suppresses mHtt effects and Atx2 effects depend on dfmr1. Atx2 knockdown reduces the cAMP response binding protein A (CrebA) transcript at dawn. CrebA transcript level shows a prominent diurnal regulation in clock neurons. Loss of CrebA also partially suppresses mHtt effects on behavior and cell loss and restoration of CrebA can suppress Atx2 effects. Our results indicate a prominent role of Atx2 in mediating mHtt pathology, specifically via its regulation of CrebA, defining a novel molecular pathway in HD pathogenesis. Circadian clocks evolved to anticipate 24 h environmental rhythms driven by the earth’s daily rotation and regulate nearly all aspects of behavior, physiology and the genome. Disruptions of the circadian clock have been associated with a wide range of human diseases, including neurodegenerative diseases such as Huntington’s disease (HD). Using an HD animal model in which a mutant Huntingtin (mHtt) protein is expressed, we identify a role for the RNA binding protein and neurodegenerative disease gene Ataxin-2 (Atx2) in mediating mHtt effects on circadian behavioral rhythms. Using transcriptomics, we identify the transcription factor CrebA as a potential target of both Atx2 and the circadian clock. Finally, we demonstrate a role for CrebA in mediating mHtt effects on circadian behavior, defining a novel Atx2-CrebA pathway in a neurodegenerative disease model. These studies define the molecular mechanisms by which mHtt can disrupt circadian rhythms identifying potential novel therapeutic targets for this uniformly fatal disease.
Collapse
Affiliation(s)
- Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Elzbieta Kula-Eversole
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Marta Iwanaszko
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Chunghun Lim
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
36
|
Göertz GP, van Bree JWM, Hiralal A, Fernhout BM, Steffens C, Boeren S, Visser TM, Vogels CBF, Abbo SR, Fros JJ, Koenraadt CJM, van Oers MM, Pijlman GP. Subgenomic flavivirus RNA binds the mosquito DEAD/H-box helicase ME31B and determines Zika virus transmission by Aedes aegypti. Proc Natl Acad Sci U S A 2019; 116:19136-19144. [PMID: 31488709 PMCID: PMC6754610 DOI: 10.1073/pnas.1905617116] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne flavivirus predominantly transmitted by Aedes aegypti mosquitoes and poses a global human health threat. All flaviviruses, including those that exclusively replicate in mosquitoes, produce a highly abundant, noncoding subgenomic flavivirus RNA (sfRNA) in infected cells, which implies an important function of sfRNA during mosquito infection. Currently, the role of sfRNA in flavivirus transmission by mosquitoes is not well understood. Here, we demonstrate that an sfRNA-deficient ZIKV (ZIKVΔSF1) replicates similar to wild-type ZIKV in mosquito cell culture but is severely attenuated in transmission by Ae. aegypti after an infectious blood meal, with 5% saliva-positive mosquitoes for ZIKVΔSF1 vs. 31% for ZIKV. Furthermore, viral titers in the mosquito saliva were lower for ZIKVΔSF1 as compared to ZIKV. Comparison of mosquito infection via infectious blood meals and intrathoracic injections showed that sfRNA is important for ZIKV to overcome the mosquito midgut barrier and to promote virus accumulation in the saliva. Next-generation sequencing of infected mosquitoes showed that viral small-interfering RNAs were elevated upon ZIKVΔSF1 as compared to ZIKV infection. RNA-affinity purification followed by mass spectrometry analysis uncovered that sfRNA specifically interacts with a specific set of Ae. aegypti proteins that are normally associated with RNA turnover and protein translation. The DEAD/H-box helicase ME31B showed the highest affinity for sfRNA and displayed antiviral activity against ZIKV in Ae. aegypti cells. Based on these results, we present a mechanistic model in which sfRNA sequesters ME31B to promote flavivirus replication and virion production to facilitate transmission by mosquitoes.
Collapse
Affiliation(s)
- Giel P Göertz
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Joyce W M van Bree
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Anwar Hiralal
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Bas M Fernhout
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Carmen Steffens
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, 6708 WE, Wageningen, The Netherlands
| | - Tessa M Visser
- Laboratory of Entomology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Chantal B F Vogels
- Laboratory of Entomology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510
| | - Sandra R Abbo
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Jelke J Fros
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | | | - Monique M van Oers
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, 6708 PB, Wageningen, The Netherlands;
| |
Collapse
|
37
|
Ki Y, Lim C. Sleep-promoting effects of threonine link amino acid metabolism in Drosophila neuron to GABAergic control of sleep drive. eLife 2019; 8:40593. [PMID: 31313987 PMCID: PMC6636906 DOI: 10.7554/elife.40593] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 06/27/2019] [Indexed: 01/09/2023] Open
Abstract
Emerging evidence indicates the role of amino acid metabolism in sleep regulation. Here we demonstrate sleep-promoting effects of dietary threonine (SPET) in Drosophila. Dietary threonine markedly increased daily sleep amount and decreased the latency to sleep onset in a dose-dependent manner. High levels of synaptic GABA or pharmacological activation of metabotropic GABA receptors (GABAB-R) suppressed SPET. By contrast, synaptic blockade of GABAergic neurons or transgenic depletion of GABAB-R in the ellipsoid body R2 neurons enhanced sleep drive non-additively with SPET. Dietary threonine reduced GABA levels, weakened metabotropic GABA responses in R2 neurons, and ameliorated memory deficits in plasticity mutants. Moreover, genetic elevation of neuronal threonine levels was sufficient for facilitating sleep onset. Taken together, these data define threonine as a physiologically relevant, sleep-promoting molecule that may intimately link neuronal metabolism of amino acids to GABAergic control of sleep drive via the neuronal substrate of sleep homeostasis. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Yoonhee Ki
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
38
|
PERIOD-controlled deadenylation of the timeless transcript in the Drosophila circadian clock. Proc Natl Acad Sci U S A 2019; 116:5721-5726. [PMID: 30833404 DOI: 10.1073/pnas.1814418116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Drosophila circadian oscillator relies on a negative transcriptional feedback loop, in which the PERIOD (PER) and TIMELESS (TIM) proteins repress the expression of their own gene by inhibiting the activity of the CLOCK (CLK) and CYCLE (CYC) transcription factors. A series of posttranslational modifications contribute to the oscillations of the PER and TIM proteins but few posttranscriptional mechanisms have been described that affect mRNA stability. Here we report that down-regulation of the POP2 deadenylase, a key component of the CCR4-NOT deadenylation complex, alters behavioral rhythms. Down-regulating POP2 specifically increases TIM protein and tim mRNA but not tim pre-mRNA, supporting a posttranscriptional role. Indeed, reduced POP2 levels induce a lengthening of tim mRNA poly(A) tail. Surprisingly, such effects are lost in per 0 mutants, supporting a PER-dependent inhibition of tim mRNA deadenylation by POP2. We report a deadenylation mechanism that controls the oscillations of a core clock gene transcript.
Collapse
|
39
|
Cusumano P, Damulewicz M, Carbognin E, Caccin L, Puricella A, Specchia V, Bozzetti MP, Costa R, Mazzotta GM. The RNA Helicase BELLE Is Involved in Circadian Rhythmicity and in Transposons Regulation in Drosophila melanogaster. Front Physiol 2019; 10:133. [PMID: 30842743 PMCID: PMC6392097 DOI: 10.3389/fphys.2019.00133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/04/2019] [Indexed: 02/05/2023] Open
Abstract
Circadian clocks control and synchronize biological rhythms of several behavioral and physiological phenomena in most, if not all, organisms. Rhythm generation relies on molecular auto-regulatory oscillations of interlocked transcriptional-translational feedback loops. Rhythmic clock-gene expression is at the base of rhythmic protein accumulation, though post-transcriptional and post-translational mechanisms have evolved to adjust and consolidate the proper pace of the clock. In Drosophila, BELLE, a conserved DEAD-box RNA helicase playing important roles in reproductive capacity, is involved in the small RNA-mediated regulation associated to the piRNA pathway. Here, we report that BELLE is implicated in the circadian rhythmicity and in the regulation of endogenous transposable elements (TEs) in both nervous system and gonads. We suggest that BELLE acts as important element in the piRNA-mediated regulation of the TEs and raise the hypothesis that this specific regulation could represent another level of post-transcriptional control adopted by the clock to ensure the proper rhythmicity.
Collapse
Affiliation(s)
- Paola Cusumano
- Department of Biology, University of Padua, Padua, Italy
| | - Milena Damulewicz
- Department of Cell Biology and Imaging, Jagiellonian University, Kraków, Poland
| | | | - Laura Caccin
- Department of Biology, University of Padua, Padua, Italy
| | - Antonietta Puricella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Valeria Specchia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Maria Pia Bozzetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Rodolfo Costa
- Department of Biology, University of Padua, Padua, Italy
| | | |
Collapse
|
40
|
Composition of the Survival Motor Neuron (SMN) Complex in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2019; 9:491-503. [PMID: 30563832 PMCID: PMC6385987 DOI: 10.1534/g3.118.200874] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Spinal Muscular Atrophy (SMA) is caused by homozygous mutations in the human survival motor neuron 1 (SMN1) gene. SMN protein has a well-characterized role in the biogenesis of small nuclear ribonucleoproteins (snRNPs), core components of the spliceosome. SMN is part of an oligomeric complex with core binding partners, collectively called Gemins. Biochemical and cell biological studies demonstrate that certain Gemins are required for proper snRNP assembly and transport. However, the precise functions of most Gemins are unknown. To gain a deeper understanding of the SMN complex in the context of metazoan evolution, we investigated its composition in Drosophila melanogaster Using transgenic flies that exclusively express Flag-tagged SMN from its native promoter, we previously found that Gemin2, Gemin3, Gemin5, and all nine classical Sm proteins, including Lsm10 and Lsm11, co-purify with SMN. Here, we show that CG2941 is also highly enriched in the pulldown. Reciprocal co-immunoprecipitation reveals that epitope-tagged CG2941 interacts with endogenous SMN in Schneider2 cells. Bioinformatic comparisons show that CG2941 shares sequence and structural similarity with metazoan Gemin4. Additional analysis shows that three other genes (CG14164, CG31950 and CG2371) are not orthologous to Gemins 6-7-8, respectively, as previously suggested. In D.melanogaster, CG2941 is located within an evolutionarily recent genomic triplication with two other nearly identical paralogous genes (CG32783 and CG32786). RNAi-mediated knockdown of CG2941 and its two close paralogs reveals that Gemin4 is essential for organismal viability.
Collapse
|
41
|
A genetic mosaic screen identifies genes modulating Notch signaling in Drosophila. PLoS One 2018; 13:e0203781. [PMID: 30235233 PMCID: PMC6147428 DOI: 10.1371/journal.pone.0203781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023] Open
Abstract
Notch signaling is conserved in most multicellular organisms and plays critical roles during animal development. The core components and major signal transduction mechanism of Notch signaling have been extensively studied. However, our understanding of how Notch signaling activity is regulated in diverse developmental processes still remains incomplete. Here, we report a genetic mosaic screen in Drosophila melanogaster that leads to identification of Notch signali ng modulators during wing development. We discovered a group of genes required for the formation of the fly wing margin, a developmental process that is strictly dependent on the balanced Notch signaling activity. These genes encode transcription factors, protein phosphatases, vacuolar ATPases and factors required for RNA transport, stability, and translation. Our data support the view that Notch signaling is controlled through a wide range of molecular processes. These results also provide foundations for further study by showing that Me31B and Wdr62 function as two novel modulators of Notch signaling activity.
Collapse
|
42
|
Lee J, Kim M, Itoh TQ, Lim C. Ataxin-2: A versatile posttranscriptional regulator and its implication in neural function. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1488. [PMID: 29869836 DOI: 10.1002/wrna.1488] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Ataxin-2 (ATXN2) is a eukaryotic RNA-binding protein that is conserved from yeast to human. Genetic expansion of a poly-glutamine tract in human ATXN2 has been implicated in several neurodegenerative diseases, likely acting through gain-of-function effects. Emerging evidence, however, suggests that ATXN2 plays more direct roles in neural function via specific molecular and cellular pathways. ATXN2 and its associated protein complex control distinct steps in posttranscriptional gene expression, including poly-A tailing, RNA stabilization, microRNA-dependent gene silencing, and translational activation. Specific RNA substrates have been identified for the functions of ATXN2 in aspects of neural physiology, such as circadian rhythms and olfactory habituation. Genetic models of ATXN2 loss-of-function have further revealed its significance in stress-induced cytoplasmic granules, mechanistic target of rapamycin signaling, and cellular metabolism, all of which are crucial for neural homeostasis. Accordingly, we propose that molecular evolution has been selecting the ATXN2 protein complex as an important trans-acting module for the posttranscriptional control of diverse neural functions. This explains how ATXN2 intimately interacts with various neurodegenerative disease genes, and suggests that loss-of-function effects of ATXN2 could be therapeutic targets for ATXN2-related neurological disorders. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Jongbo Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Minjong Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Taichi Q Itoh
- Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| |
Collapse
|
43
|
High-Amplitude Circadian Rhythms in Drosophila Driven by Calcineurin-Mediated Post-translational Control of sarah. Genetics 2018; 209:815-828. [PMID: 29724861 DOI: 10.1534/genetics.118.300808] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 12/26/2022] Open
Abstract
Post-translational control is a crucial mechanism for circadian timekeeping. Evolutionarily conserved kinases and phosphatases have been implicated in circadian phosphorylation and the degradation of clock-relevant proteins, which sustain high-amplitude rhythms with 24-hr periodicity in animal behaviors and physiology. Here, we report a novel clock function of the heterodimeric Ca2+/calmodulin-dependent phosphatase calcineurin and its regulator sarah (sra) in Drosophila Genomic deletion of the sra locus dampened circadian locomotor activity rhythms in free-running constant dark after entrainment in light-dark cycles. Poor rhythms in sra mutant behaviors were accompanied by lower expression of two oscillating clock proteins, PERIOD (PER) and TIMELESS (TIM), at the post-transcriptional level. RNA interference-mediated sra depletion in circadian pacemaker neurons was sufficient to phenocopy loss-of-function mutation in sra On the other hand, a constitutively active form of the catalytic calcineurin subunit, Pp2B-14DACT, shortened circadian periodicity in locomotor behaviors and phase-advanced PER and TIM rhythms when overexpressed in clock neurons. Heterozygous sra deletion induced behavioral arrhythmicity in Pp2B-14DACT flies, whereas sra overexpression rescued short periods in these animals. Finally, pharmacological inhibition of calcineurin in either wild-type flies or clock-less S2 cells decreased the levels of PER and TIM, likely by facilitating their proteasomal degradation. Taken together, these data suggest that sra negatively regulates calcineurin by cell-autonomously titrating calcineurin-dependent stabilization of PER and TIM proteins, thereby sustaining high-amplitude behavioral rhythms in Drosophila.
Collapse
|
44
|
DeHaan H, McCambridge A, Armstrong B, Cruse C, Solanki D, Trinidad JC, Arkov AL, Gao M. An in vivo proteomic analysis of the Me31B interactome in Drosophila germ granules. FEBS Lett 2017; 591:3536-3547. [PMID: 28945271 DOI: 10.1002/1873-3468.12854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/11/2022]
Abstract
Drosophila Me31B is a conserved protein of germ granules, ribonucleoprotein complexes essential for germ cell development. Me31B post-transcriptionally regulates mRNAs by interacting with other germ granule proteins. However, a Me31B interactome is lacking. Here, we use an in vivo proteomics approach to show that the Me31B interactome contains polypeptides from four functional groups: RNA regulatory proteins, glycolytic enzymes, cytoskeleton/motor proteins, and germ plasm components. We further show that Me31B likely colocalizes with the germ plasm components Tudor (Tud), Vasa, and Aubergine in the nuage and germ plasm and provide evidence that Me31B may directly bind to Tud in a symmetrically dimethylated arginine-dependent manner. Our study supports the role of Me31B in RNA regulation and suggests its novel roles in germ granule assembly and function.
Collapse
Affiliation(s)
- Hunter DeHaan
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | | | | | - Carlie Cruse
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | - Dhruv Solanki
- Biology Department, Indiana University Northwest, Gary, IN, USA
| | | | - Alexey L Arkov
- Department of Biological Sciences, Murray State University, Murray, KY, USA
| | - Ming Gao
- Biology Department, Indiana University Northwest, Gary, IN, USA
| |
Collapse
|
45
|
Rhythmic Behavior Is Controlled by the SRm160 Splicing Factor in Drosophila melanogaster. Genetics 2017; 207:593-607. [PMID: 28801530 DOI: 10.1534/genetics.117.300139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023] Open
Abstract
Circadian clocks organize the metabolism, physiology, and behavior of organisms throughout the day-night cycle by controlling daily rhythms in gene expression at the transcriptional and post-transcriptional levels. While many transcription factors underlying circadian oscillations are known, the splicing factors that modulate these rhythms remain largely unexplored. A genome-wide assessment of the alterations of gene expression in a null mutant of the alternative splicing regulator SR-related matrix protein of 160 kDa (SRm160) revealed the extent to which alternative splicing impacts on behavior-related genes. We show that SRm160 affects gene expression in pacemaker neurons of the Drosophila brain to ensure proper oscillations of the molecular clock. A reduced level of SRm160 in adult pacemaker neurons impairs circadian rhythms in locomotor behavior, and this phenotype is caused, at least in part, by a marked reduction in period (per) levels. Moreover, rhythmic accumulation of the neuropeptide PIGMENT DISPERSING FACTOR in the dorsal projections of these neurons is abolished after SRm160 depletion. The lack of rhythmicity in SRm160-downregulated flies is reversed by a fully spliced per construct, but not by an extra copy of the endogenous locus, showing that SRm160 positively regulates per levels in a splicing-dependent manner. Our findings highlight the significant effect of alternative splicing on the nervous system and particularly on brain function in an in vivo model.
Collapse
|
46
|
Auburger G, Sen NE, Meierhofer D, Başak AN, Gitler AD. Efficient Prevention of Neurodegenerative Diseases by Depletion of Starvation Response Factor Ataxin-2. Trends Neurosci 2017; 40:507-516. [DOI: 10.1016/j.tins.2017.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/09/2017] [Indexed: 12/13/2022]
|