1
|
Ghavami S, Zamani M, Ahmadi M, Erfani M, Dastghaib S, Darbandi M, Darbandi S, Vakili O, Siri M, Grabarek BO, Boroń D, Zarghooni M, Wiechec E, Mokarram P. Epigenetic regulation of autophagy in gastrointestinal cancers. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166512. [PMID: 35931405 DOI: 10.1016/j.bbadis.2022.166512] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 11/09/2022]
Abstract
The development of novel therapeutic approaches is necessary to manage gastrointestinal cancers (GICs). Considering the effective molecular mechanisms involved in tumor growth, the therapeutic response is pivotal in this process. Autophagy is a highly conserved catabolic process that acts as a double-edged sword in tumorigenesis and tumor inhibition in a context-dependent manner. Depending on the stage of malignancy and cellular origin of the tumor, autophagy might result in cancer cell survival or death during the GICs' progression. Moreover, autophagy can prevent the progression of GIC in the early stages but leads to chemoresistance in advanced stages. Therefore, targeting specific arms of autophagy could be a promising strategy in the prevention of chemoresistance and treatment of GIC. It has been revealed that autophagy is a cytoplasmic event that is subject to transcriptional and epigenetic regulation inside the nucleus. The effect of epigenetic regulation (including DNA methylation, histone modification, and expression of non-coding RNAs (ncRNAs) in cellular fate is still not completely understood. Recent findings have indicated that epigenetic alterations can modify several genes and modulators, eventually leading to inhibition or promotion of autophagy in different cancer stages, and mediating chemoresistance or chemosensitivity. The current review focuses on the links between autophagy and epigenetics in GICs and discusses: 1) How autophagy and epigenetics are linked in GICs, by considering different epigenetic mechanisms; 2) how epigenetics may be involved in the alteration of cancer-related phenotypes, including cell proliferation, invasion, and migration; and 3) how epidrugs modulate autophagy in GICs to overcome chemoresistance.
Collapse
Affiliation(s)
- Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institute of Hematology and Oncology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland.
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran
| | - Mehran Erfani
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Darbandi
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran; Gene Therapy and Regenerative Medicine Research Center, Hope Generation Foundation, Tehran, Iran
| | - Sara Darbandi
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran; Gene Therapy and Regenerative Medicine Research Center, Hope Generation Foundation, Tehran, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Morvarid Siri
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Beniamin Oskar Grabarek
- Department of Histology, Cytophysiology, and Embryology in Zabrze, Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland; Department of Gynecology and Obstetrics in Zabrze, Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Dariusz Boroń
- Department of Histology, Cytophysiology, and Embryology in Zabrze, Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland; Department of Gynecology and Obstetrics in Zabrze, Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Maryam Zarghooni
- Department of Laboratory Medicine and Pathobiology, University of Toronto Alumni, Toronto, Canada
| | - Emilia Wiechec
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Castelli LM, Benson BC, Huang WP, Lin YH, Hautbergue GM. RNA Helicases in Microsatellite Repeat Expansion Disorders and Neurodegeneration. Front Genet 2022; 13:886563. [PMID: 35646086 PMCID: PMC9133428 DOI: 10.3389/fgene.2022.886563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Short repeated sequences of 3-6 nucleotides are causing a growing number of over 50 microsatellite expansion disorders, which mainly present with neurodegenerative features. Although considered rare diseases in relation to the relatively low number of cases, these primarily adult-onset conditions, often debilitating and fatal in absence of a cure, collectively pose a large burden on healthcare systems in an ageing world population. The pathological mechanisms driving disease onset are complex implicating several non-exclusive mechanisms of neuronal injury linked to RNA and protein toxic gain- and loss- of functions. Adding to the complexity of pathogenesis, microsatellite repeat expansions are polymorphic and found in coding as well as in non-coding regions of genes. They form secondary and tertiary structures involving G-quadruplexes and atypical helices in repeated GC-rich sequences. Unwinding of these structures by RNA helicases plays multiple roles in the expression of genes including repeat-associated non-AUG (RAN) translation of polymeric-repeat proteins with aggregating and cytotoxic properties. Here, we will briefly review the pathogenic mechanisms mediated by microsatellite repeat expansions prior to focus on the RNA helicases eIF4A, DDX3X and DHX36 which act as modifiers of RAN translation in C9ORF72-linked amyotrophic lateral sclerosis/frontotemporal dementia (C9ORF72-ALS/FTD) and Fragile X-associated tremor/ataxia syndrome (FXTAS). We will further review the RNA helicases DDX5/17, DHX9, Dicer and UPF1 which play additional roles in the dysregulation of RNA metabolism in repeat expansion disorders. In addition, we will contrast these with the roles of other RNA helicases such as DDX19/20, senataxin and others which have been associated with neurodegeneration independently of microsatellite repeat expansions. Finally, we will discuss the challenges and potential opportunities that are associated with the targeting of RNA helicases for the development of future therapeutic approaches.
Collapse
Affiliation(s)
- Lydia M Castelli
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Bridget C Benson
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Wan-Ping Huang
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Ya-Hui Lin
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
3
|
Wang J, Li T, Deng S, Ma E, Zhang J, Xing S. The RNA helicase DDX3 is required for ovarian development and oocyte maturation in Locusta migratoria. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2021; 106:e21775. [PMID: 33644918 DOI: 10.1002/arch.21775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 06/12/2023]
Abstract
DDX3 represents a well-defined subfamily of DEAD-box RNA helicase and exerts multiple functions in RNA metabolism, cell cycle, tumorigenesis, signal pathway, and fertility. Our previous study has shown that LmDDX3, the ortholog of DDX3 in Locusta migratoria, is ubiquitously expressed, and with a high abundance in testis and ovary. Knockdown of LmDDX3 results in a lethal phenotype in nymph, but it still remains unclear for its role in reproductive process. In this study, we therefore characterized LmDDX3 expression in female adult locust and analyzed its function in oocyte development. LmDDX3 was expressed in all tissues examined with significant more transcripts in ovary and hindgut. In ovary, a strong expression level was detected at the day just after adult eclosion, and a dramatic reduction then occurred during the oocyte development. LmDDX3 RNAi led to a reduced vitellogenin (Vg) expression in fat body via partially at least, the JH signaling pathway, and caused an upregulation of vitellogenin receptor (VgR) in ovary, and thus blocked the ovarian development and oocyte maturation. Sequence and phylogenetic analysis indicated that LmDDX3 was closely related to termite DDX3. Taken together, these data reveal a critical role for LmDDX3 in regulating the transcription of Vg and VgR, two major factors in vitellogenesis that is a key process required for ovary development and oocyte maturation in locust, and contribute thereof a new putative target for locust biological control.
Collapse
Affiliation(s)
- Junxiu Wang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- College of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Tingting Li
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- College of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Sufang Deng
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- College of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Enbo Ma
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Agricultural Integrated Pest Management, Taiyuan, Shanxi, China
| | - Jianzhen Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Agricultural Integrated Pest Management, Taiyuan, Shanxi, China
| | - Shuping Xing
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Agricultural Integrated Pest Management, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Zhang M, Chen Y, Cheng X, Cai Z, Qiu S. GATA1/SP1 and miR-874 mediate enterovirus-71-induced apoptosis in a granzyme-B-dependent manner in Jurkat cells. Arch Virol 2020; 165:2531-2540. [PMID: 32851429 DOI: 10.1007/s00705-020-04783-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/21/2020] [Indexed: 01/20/2023]
Abstract
Enterovirus 71 (EV71)-induced T lymphocyte apoptosis plays an important role in hand, foot, and mouth disease (HFMD), and granzyme B (GZMB) has been shown to be critical for this process. However, the mechanisms underlying GZMB-mediated apoptosis of T lymphocytes remain unknown. In this study, we investigated whether transcription factors and microRNAs (miRNAs) are involved in GZMB-mediated apoptosis of T lymphocytes in response to EV71 infection. Our findings indicated that EV71 infection significantly induced apoptosis in Jurkat cells, a human T lymphocytes cell line, as revealed in flow cytometric analysis. Furthermore, EV71 increased the expression of pro-apoptosis Bcl-2-associated X (Bax) and cleaved caspase 3 but decreased the expression of anti-apoptosis B-cell lymphoma protein 2 (Bcl2). GZMB knockdown decreased cell apoptosis and prevented EV71-induced changes in the expression of Bax, cleaved caspase 3, and Bcl2 in Jurkat cells, highlighting the role of GZMB as a key factor in EV71-induced apoptosis. Our study also indicated that overexpression of the transcription factors GATA binding factor 1 (GATA1) and specificity protein 1 (SP1) significantly increased luciferase activity when this gene was inserted in the GZMB 3' untranslated region (3'UTR). GATA1/SP1 overexpression induced cell apoptosis, increased the expression of Bax and cleaved caspase 3, and decreased the expression of Bcl2. Finally, our results suggested that miR-874 plays an essential role in GZMB-mediated cell apoptosis, since an miR-874 mimic decreases the expression of GZMB by targeting its 3'UTR. Collectively, these data indicated that GATA1/SP1 and miR-874 mediate EV71-induced apoptosis in a granzyme B-dependent manner. This signaling pathway may provide a new pharmacological target for the prevention and treatment of HFMD.
Collapse
Affiliation(s)
- Meijuan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Ying Chen
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiangjun Cheng
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhenzhen Cai
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Shengfeng Qiu
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
5
|
The Drosophila RNA Helicase Belle (DDX3) Non-Autonomously Suppresses Germline Tumorigenesis Via Regulation of a Specific mRNA Set. Cells 2020; 9:cells9030550. [PMID: 32111103 PMCID: PMC7140462 DOI: 10.3390/cells9030550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/28/2022] Open
Abstract
DDX3 subfamily DEAD-box RNA helicases are essential developmental regulators of RNA metabolism in eukaryotes. belle, the single DDX3 ortholog in Drosophila, is required for fly viability, fertility, and germline stem cell maintenance. Belle is involved both in translational activation and repression of target mRNAs in different tissues; however, direct targets of Belle in the testes are essentially unknown. Here we showed that belle RNAi knockdown in testis cyst cells caused a disruption of adhesion between germ and cyst cells and generation of tumor-like clusters of stem-like germ cells. Ectopic expression of β-integrin in cyst cells rescued early stages of spermatogenesis in belle knockdown testes, indicating that integrin adhesion complexes are required for the interaction between somatic and germ cells in a cyst. To address Belle functions in spermatogenesis in detail we performed cross-linking immunoprecipitation and sequencing (CLIP-seq) analysis and identified multiple mRNAs that interacted with Belle in the testes. The set of Belle targets includes transcripts of proteins that are essential for preventing the tumor-like clusters of germ cells and for sustaining spermatogenesis. By our hypothesis, failures in the translation of a number of mRNA targets additively contribute to developmental defects observed in the testes with belle knockdowns both in cyst cells and in the germline.
Collapse
|
6
|
Dhamodaran K, Subramani M, Krishna L, Matalia H, Jayadev C, Chinnappaiah N, Shetty R, Das D. Temporal Regulation of Notch Signaling and Its Influence on the Differentiation of Ex Vivo Cultured Limbal Epithelial Cells. Curr Eye Res 2019; 45:459-470. [PMID: 31558050 DOI: 10.1080/02713683.2019.1673436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Purpose: Notch signaling plays a vital role in the differentiation and proliferation of corneal epithelial cells from limbal stem cells. The temporal regulation of Notch signaling during this differentiation remains unknown. Hence, we investigated the importance of temporal activation/blockage of Notch signaling during corneal differentiation.Methods: Human limbal epithelial cultures were established with and without Notch activators (rec-Human Jagged1 Fc chimera) and pharmacological blockers (LY-411575). The modulation of Notch signaling was done at different time points during cell differentiation, which were collected on Day 14 for further analysis of differentiation, proliferation, maturation and apoptosis using RT-qPCR and immunofluorescence staining.Results: The activation of Notch signaling at Day 8 resulted in the highest number of mature corneal epithelial cells (p = .008) and pro-apoptosis marker BAX (p = .0001) with no increase in the number of corneal progenitors, and proliferation marker Ki67 compared to untreated controls. Cultures grown in the presence of Notch signaling blockers showed a significantly higher number of corneal progenitors (p = .0001) and proliferation marker Ki67 (p = .02) but lower corneal epithelial marker CK3/CK12 (p = .0007) and no difference in the pro-apoptotic marker BAX compared to untreated controls.Conclusion: During the differentiation of limbal epithelial cells to the corneal epithelial cell type, Day 8 seems to be a crucial window to modulate Notch signaling for a customized outcome.
Collapse
Affiliation(s)
- Kamesh Dhamodaran
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Murali Subramani
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Lekshmi Krishna
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Himanshu Matalia
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Chaitra Jayadev
- Department of Vitreo-retinal services, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Nandini Chinnappaiah
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Debashish Das
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| |
Collapse
|
7
|
Knapp EM, Li W, Sun J. Downregulation of homeodomain protein Cut is essential for Drosophila follicle maturation and ovulation. Development 2019; 146:dev179002. [PMID: 31444217 PMCID: PMC6765176 DOI: 10.1242/dev.179002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/19/2019] [Indexed: 01/19/2023]
Abstract
Proper development and maturation of a follicle is essential for successful ovulation and reproduction; however, the molecular mechanisms for follicle maturation, particularly for somatic follicle cell differentiation, are poorly understood. During Drosophila oogenesis, the somatic follicle cells encasing oocytes undergo two distinct well-established transitions: the mitotic to endocycle switch at stage 6/7 and the endocycle to gene amplification switch at stage10A/10B. Here, we identify a novel third follicle cell transition that occurs in the final stages of oogenesis (stage 13/14). This late follicle cell transition is characterized by upregulation of the transcription factor Hindsight (Hnt), and downregulation of the homeodomain transcription factor Cut and the zinc-finger transcription factor Tramtrack-69 (Ttk69). We demonstrate that inducing expression of Cut in stage 14 follicle cells is sufficient to inhibit follicle rupture and ovulation through its negative regulation of Hnt and promotion of Ttk69 expression. Our work illustrates the importance of the stage13/14 transition for follicle maturation and demonstrates the complex regulation required for somatic follicle cells to differentiate into a state primed for follicle rupture and ovulation.
Collapse
Affiliation(s)
- Elizabeth M Knapp
- Department of Physiology & Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Wei Li
- Department of Physiology & Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Jianjun Sun
- Department of Physiology & Neurobiology, University of Connecticut, Storrs, CT 06269, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
8
|
Linsalata AE, He F, Malik AM, Glineburg MR, Green KM, Natla S, Flores BN, Krans A, Archbold HC, Fedak SJ, Barmada SJ, Todd PK. DDX3X and specific initiation factors modulate FMR1 repeat-associated non-AUG-initiated translation. EMBO Rep 2019; 20:e47498. [PMID: 31347257 PMCID: PMC6726903 DOI: 10.15252/embr.201847498] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
A CGG trinucleotide repeat expansion in the 5' UTR of FMR1 causes the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). This repeat supports a non-canonical mode of protein synthesis known as repeat-associated, non-AUG (RAN) translation. The mechanism underlying RAN translation at CGG repeats remains unclear. To identify modifiers of RAN translation and potential therapeutic targets, we performed a candidate-based screen of eukaryotic initiation factors and RNA helicases in cell-based assays and a Drosophila melanogaster model of FXTAS. We identified multiple modifiers of toxicity and RAN translation from an expanded CGG repeat in the context of the FMR1 5'UTR. These include the DEAD-box RNA helicase belle/DDX3X, the helicase accessory factors EIF4B/4H, and the start codon selectivity factors EIF1 and EIF5. Disrupting belle/DDX3X selectively inhibited FMR1 RAN translation in Drosophila in vivo and cultured human cells, and mitigated repeat-induced toxicity in Drosophila and primary rodent neurons. These findings implicate RNA secondary structure and start codon fidelity as critical elements mediating FMR1 RAN translation and identify potential targets for treating repeat-associated neurodegeneration.
Collapse
Affiliation(s)
- Alexander E Linsalata
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Fang He
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Department of Biological and Health SciencesTexas A&M University, KingsvilleKingsvilleTXUSA
| | - Ahmed M Malik
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMIUSA
| | | | - Katelyn M Green
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Sam Natla
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Brittany N Flores
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Amy Krans
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | | | | | - Sami J Barmada
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Peter K Todd
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Ann Arbor VA Medical CenterAnn ArborMIUSA
| |
Collapse
|
9
|
Liao SE, Kandasamy SK, Zhu L, Fukunaga R. DEAD-box RNA helicase Belle posttranscriptionally promotes gene expression in an ATPase activity-dependent manner. RNA (NEW YORK, N.Y.) 2019; 25:825-839. [PMID: 30979781 PMCID: PMC6573787 DOI: 10.1261/rna.070268.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
Drosophila Belle (human ortholog DDX3) is a conserved DEAD-box RNA helicase implicated in regulating gene expression. However, the molecular mechanisms by which Belle/DDX3 regulates gene expression are poorly understood. Here we performed systematic mutational analysis to determine the contributions of conserved motifs within Belle to its in vivo function. We found that Belle RNA-binding and RNA-unwinding activities and intrinsically disordered regions (IDRs) are required for Belle in vivo function. Expression of Belle ATPase mutants that cannot bind, hydrolyze, or release ATP resulted in dominant toxic phenotypes. Mechanistically, we discovered that Belle up-regulates reporter protein level when tethered to reporter mRNA, without corresponding changes at the mRNA level, indicating that Belle promotes translation of mRNA that it binds. Belle ATPase activity and amino-terminal IDR were required for this translational promotion activity. We also found that ectopic ovary expression of dominant Belle ATPase mutants decreases levels of cyclin proteins, including Cyclin B, without corresponding changes in their mRNA levels. Finally, we found that Belle binds endogenous cyclin B mRNA. We propose that Belle promotes translation of specific target mRNAs, including cyclin B mRNA, in an ATPase activity-dependent manner.
Collapse
Affiliation(s)
- Susan E Liao
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Suresh K Kandasamy
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Li Zhu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
10
|
John AA, Prakash R, Singh D. miR-487b-3p impairs osteoblastogenesis by targeting Notch-regulated ankyrin-repeat protein (Nrarp). J Endocrinol 2019; 241:249-263. [PMID: 30978699 DOI: 10.1530/joe-19-0015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Abstract
miRNAs have appeared as critical controllers of gene expression at post-transcriptional level either by degrading RNA transcripts or repressing translation. It is evident from the ever-growing scientific literature that miRNAs play a significant role in osteoblast commitment and differentiation. Here, we report that overexpression of miR-487b-3p leads to inhibition of osteoblastic differentiation. Using in silico approaches, Nrarp was found to be the direct target of miR-487b-3p, which was further validated by luciferase 3' UTR reporter assay. Nrarp inhibits Notch-1 signaling and promotes Wnt signaling by stabilization of LEF-1. Role of miR-487b-3p in regulating canonical Wnt and Notch signaling was determined by western blotting. Protein levels of Nrarp, RUNX-2, Lef1 and β catenin were reduced in osteoblasts cells transfected with miR-487b-3p, whereas protein levels of Notch1, Hes1 and P-β catenin were upregulated when osteoblast cells were transfected with miR-487b-3p. These outcomes were reversed after treating cells with anti-miR-487b-3p. Further silencing of miR-487b-3p in neonatal Balb/c mice attenuated all the inhibitory actions of miR-487b-3p on osteoblast differentiation. Importantly, in vivo action of anti-miR-487b-3p to ovariectomized osteopenic BALB/c mice steered to significant enhancement in trabecular bone microarchitecture. Furthermore, the bio-mechanical properties of isolated femurs were enhanced in anti-miR-487b-3p-treated mice. Overall, miR-487b-3p negatively regulates osteogenesis by suppressing Nrarp expression, which in turn, suppresses Runx-2 and Wnt signaling, both of which play a pivotal action in osteoblast differentiation.
Collapse
Affiliation(s)
- Aijaz A John
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
| | - Divya Singh
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
11
|
Fic W, Faria C, St Johnston D. IMP regulates Kuzbanian to control the timing of Notch signalling in Drosophila follicle cells. Development 2019; 146:dev.168963. [PMID: 30635283 PMCID: PMC6361131 DOI: 10.1242/dev.168963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022]
Abstract
The timing of Drosophila egg chamber development is controlled by a germline Delta signal that activates Notch in the follicle cells to induce them to cease proliferation and differentiate. Here, we report that follicle cells lacking the RNA-binding protein IMP go through one extra division owing to a delay in the Delta-dependent S2 cleavage of Notch. The timing of Notch activation has previously been shown to be controlled by cis-inhibition by Delta in the follicle cells, which is relieved when the miRNA pathway represses Delta expression. imp mutants are epistatic to Delta mutants and give an additive phenotype with belle and Dicer-1 mutants, indicating that IMP functions independently of both cis-inhibition and the miRNA pathway. We find that the imp phenotype is rescued by overexpression of Kuzbanian, the metalloprotease that mediates the Notch S2 cleavage. Furthermore, Kuzbanian is not enriched at the apical membrane in imp mutants, accumulating instead in late endosomes. Thus, IMP regulates Notch signalling by controlling the localisation of Kuzbanian to the apical domain, where Notch cleavage occurs, revealing a novel regulatory step in the Notch pathway.
Collapse
Affiliation(s)
| | | | - Daniel St Johnston
- The Gurdon Institute and The Department of Genetics, University of Cambridge, Tennis Court Rd, Cambridge CB2 1QN, UK
| |
Collapse
|
12
|
Jenny FH, Basler K. Drosophila DDX3/Belle Exerts Its Function Outside of the Wnt/Wingless Signaling Pathway. PLoS One 2016; 11:e0166862. [PMID: 28030561 PMCID: PMC5193393 DOI: 10.1371/journal.pone.0166862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 11/04/2016] [Indexed: 11/19/2022] Open
Abstract
The helicases human DDX3 and Drosophila Belle (Bel) are part of a well-defined subfamily of the DEAD-box helicases. Individual subfamily-members perform a myriad of functions in nuclear and cytosolic RNA metabolism. It has also been reported that DDX3X is involved in cell signaling, including IFN-α and IFN-β inducing pathways upon viral infection as well as in Wnt signaling. Here we used a collection of EMS-induced bel alleles recovered from a Wingless (Wg) suppressor screen to analyze the role of the Drosophila homolog of DDX3 in Wg/Wnt signaling. These EMS alleles, as well as a P-element induced null allele and RNAi-mediated knock down of bel, all suppressed the phenotype of ectopic Wg signaling in the eye. However, they did not affect the expression of known Wg target genes like senseless, Distalless or wingful/Notum. Ectopic Wg signaling in eye imaginal discs induces apoptosis by increasing grim expression. Mutations in bel revert grim expression to wild-type levels. Together, these results indicate that Bel does not function as a core component in the Drosophila Wg pathway, and that mutations affecting its helicase function suppress the effects of ectopic Wg signaling downstream of the canonical pathway.
Collapse
Affiliation(s)
- Fabian H. Jenny
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Konrad Basler
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Yang H, Li M, Hu X, Xin T, Zhang S, Zhao G, Xuan T, Li M. MicroRNA-dependent roles of Drosha and Pasha in the Drosophila larval ovary morphogenesis. Dev Biol 2016; 416:312-23. [DOI: 10.1016/j.ydbio.2016.06.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 01/04/2023]
|
14
|
The multiple functions of RNA helicases as drivers and regulators of gene expression. Nat Rev Mol Cell Biol 2016; 17:426-38. [PMID: 27251421 DOI: 10.1038/nrm.2016.50] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
RNA helicases comprise the largest family of enzymes involved in the metabolism of mRNAs, the processing and fate of which rely on their packaging into messenger ribonucleoprotein particles (mRNPs). In this Review, we describe how the capacity of some RNA helicases to either remodel or lock the composition of mRNP complexes underlies their pleiotropic functions at different steps of the gene expression process. We illustrate the roles of RNA helicases in coordinating gene expression steps and programmes, and propose that RNA helicases function as molecular drivers and guides of the progression of their mRNA substrates from one RNA-processing factory to another, to a productive mRNA pool that leads to protein synthesis or to unproductive mRNA pools that are stored or degraded.
Collapse
|
15
|
Macedo LMF, Nunes FMF, Freitas FCP, Pires CV, Tanaka ED, Martins JR, Piulachs MD, Cristino AS, Pinheiro DG, Simões ZLP. MicroRNA signatures characterizing caste-independent ovarian activity in queen and worker honeybees (Apis mellifera L.). INSECT MOLECULAR BIOLOGY 2016; 25:216-26. [PMID: 26853694 DOI: 10.1111/imb.12214] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Queen and worker honeybees differ profoundly in reproductive capacity. The queen of this complex society, with 200 highly active ovarioles in each ovary, is the fertile caste, whereas the workers have approximately 20 ovarioles as a result of receiving a different diet during larval development. In a regular queenright colony, the workers have inactive ovaries and do not reproduce. However, if the queen is sensed to be absent, some of the workers activate their ovaries, producing viable haploid eggs that develop into males. Here, a deep-sequenced ovary transcriptome library of reproductive workers was used as supporting data to assess the dynamic expression of the regulatory molecules and microRNAs (miRNAs) of reproductive and nonreproductive honeybee females. In this library, most of the differentially expressed miRNAs are related to ovary physiology or oogenesis. When we quantified the dynamic expression of 19 miRNAs in the active and inactive worker ovaries and compared their expression in the ovaries of virgin and mated queens, we noted that some miRNAs (miR-1, miR-31a, miR-13b, miR-125, let-7 RNA, miR-100, miR-276, miR-12, miR-263a, miR-306, miR-317, miR-92a and miR-9a) could be used to identify reproductive and nonreproductive statuses independent of caste. Furthermore, integrative gene networks suggested that some candidate miRNAs function in the process of ovary activation in worker bees.
Collapse
Affiliation(s)
- L M F Macedo
- Departamento De Genética, Faculdade De Medicina De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| | - F M F Nunes
- Departamento De Genética E Evolução, Centro De Ciências Biológicas E Da Saúde, Universidade Federal De São Carlos, São Carlos, Brazil
| | - F C P Freitas
- Departamento De Genética, Faculdade De Medicina De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| | - C V Pires
- Departamento De Genética, Faculdade De Medicina De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| | - E D Tanaka
- Departamento De Genética, Faculdade De Medicina De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| | - J R Martins
- Departamento De Genética, Faculdade De Medicina De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| | - M-D Piulachs
- Institute of Evolutionary Biology, CSIC-Universitat Pompeu Fabra, Barcelona, Spain
| | - A S Cristino
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - D G Pinheiro
- Departamento De Tecnologia, Faculdade De Ciências Agrárias E Veterinárias, Universidade Estadual Paulista, Jaboticabal, Brazil
| | - Z L P Simões
- Departamento De Biologia, Faculdade De Filosofia, Ciências E Letras De Ribeirão Preto, Universidade De São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
16
|
Lo PK, Huang YC, Poulton JS, Leake N, Palmer WH, Vera D, Xie G, Klusza S, Deng WM. RNA helicase Belle/DDX3 regulates transgene expression in Drosophila. Dev Biol 2016; 412:57-70. [PMID: 26900887 DOI: 10.1016/j.ydbio.2016.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 11/28/2022]
Abstract
Belle (Bel), the Drosophila homolog of the yeast DEAD-box RNA helicase DED1 and human DDX3, has been shown to be required for oogenesis and female fertility. Here we report a novel role of Bel in regulating the expression of transgenes. Abrogation of Bel by mutations or RNAi induces silencing of a variety of P-element-derived transgenes. This silencing effect depends on downregulation of their RNA levels. Our genetic studies have revealed that the RNA helicase Spindle-E (Spn-E), a nuage RNA helicase that plays a crucial role in regulating RNA processing and PIWI-interacting RNA (piRNA) biogenesis in germline cells, is required for loss-of-bel-induced transgene silencing. Conversely, Bel abrogation alleviates the nuage-protein mislocalization phenotype in spn-E mutants, suggesting a competitive relationship between these two RNA helicases. Additionally, disruption of the chromatin remodeling factor Mod(mdg4) or the microRNA biogenesis enzyme Dicer-1 (Dcr-1) also alleviates the transgene-silencing phenotypes in bel mutants, suggesting the involvement of chromatin remodeling and microRNA biogenesis in loss-of-bel-induced transgene silencing. Finally we show that genetic inhibition of Bel function leads to de novo generation of piRNAs from the transgene region inserted in the genome, suggesting a potential piRNA-dependent mechanism that may mediate transgene silencing as Bel function is inhibited.
Collapse
Affiliation(s)
- Pang-Kuo Lo
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Yi-Chun Huang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - John S Poulton
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Nicholas Leake
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - William H Palmer
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Daniel Vera
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Stephen Klusza
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
17
|
Kotov AA, Olenkina OM, Kibanov MV, Olenina LV. RNA helicase Belle (DDX3) is essential for male germline stem cell maintenance and division in Drosophila. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1093-105. [PMID: 26876306 DOI: 10.1016/j.bbamcr.2016.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/19/2016] [Accepted: 02/09/2016] [Indexed: 01/08/2023]
Abstract
The present study showed that RNA helicase Belle (DDX3) was required intrinsically for mitotic progression and survival of germline stem cells (GSCs) and spermatogonial cells in the Drosophila melanogaster testes. We found that deficiency of Belle in the male germline resulted in a strong germ cell loss phenotype. Early germ cells are lost through cell death, whereas somatic hub and cyst cell populations are maintained. The observed phenotype is related to that of the human Sertoli Cell-Only Syndrome caused by the loss of DBY (DDX3) expression in the human testes and results in a complete lack of germ cells with preservation of somatic Sertoli cells. We found the hallmarks of mitotic G2 delay in early germ cells of the larval testes of bel mutants. Both mitotic cyclins, A and B, are markedly reduced in the gonads of bel mutants. Transcription levels of cycB and cycA decrease significantly in the testes of hypomorph bel mutants. Overexpression of Cyclin B in the germline partially rescues germ cell survival, mitotic progression and fertility in the bel-RNAi knockdown testes. Taken together, these results suggest that a role of Belle in GSC maintenance and regulation of early germ cell divisions is associated with the expression control of mitotic cyclins.
Collapse
Affiliation(s)
- Alexei A Kotov
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Oxana M Olenkina
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Mikhail V Kibanov
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Ludmila V Olenina
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia.
| |
Collapse
|
18
|
Lu H, He X, Wang Q, Zheng D, Han Y, Yang W, Liu T. MicroRNA let-7b-regulated epidermal stem cell proliferation in hypertrophied anal papillae. Mol Med Rep 2015; 12:4821-8. [PMID: 26133574 PMCID: PMC4581746 DOI: 10.3892/mmr.2015.4017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 04/10/2015] [Indexed: 01/09/2023] Open
Abstract
The present study investigated the role of epidermal stem cell-expressed microRNA let-7b in the pathogenesis of hypertrophied anal papillae. Hypertrophied anal papillae were examined for the presence of epidermal stem cells. Epidermal stem cells were identified using flow cytometry and immunofluorescent staining for the cell surface markers, integrin α6 and integrin β1 subunits. Expression levels of microRNA let-7b in α6+/β1+and α6-/β1-cells were compared using reverse transcription-quantitative polymerase chain reaction and northern blotting. Lentivirus-mediated expression of microRNA let-7b in epidermal stem cells was utilized in order to study the effects of this microRNA on the cell cycle proteins, cyclin D1 (CCND1) and cyclin-dependent kinase 4 (CDK4). MicroRNA let-7b-overexpressing cells were examined using flow cytometry, in order to determine the effects of the microRNA on cell cycle progression. α6+/β1+epidermal stem cells were identified in hypertrophic anal papillae. Following isolation and enrichment of the α6+/β1+population, these cells were found to have a rapid rate of proliferation in vitro. The expression of cell cycle-related proteins was elevated in this population, compared with that in α6-/β1-cells. The expression of microRNA let-7b in α6+/β1+epidermal stem cells was significantly lower than that in α6-/β1-cells. Two microRNA let-7b target genes, CCND1 and CDK4, were found to be upregulated in α6+/β1+cells. When the exogenous precursor, microRNA let-7, was overexpressed in α6+/β1+ epidermal stem cells, the cell proliferation rate was significantly lower than that in cells expressing microRNA let-7 containing a mutated seed sequence. The addition of exogenous microRNA let-7 resulted in an increased expression level of mature microRNA let-7b, while the expression of CCND1 and CDK4 was reduced. Epidermal stem cells transfected with microRNA let-7b were arrested in the G2/M phase and the percentage of cells in S-phase was significantly reduced. In conclusion, let-7b expression results in upregulation of the cell cycle-related proteins, CCND1 and CDK4, resulting in the excessive proliferation that leads to the formation of hypertrophic anal papillae.
Collapse
Affiliation(s)
- Hong Lu
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xiang He
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Qingming Wang
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - De Zheng
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ye Han
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Wei Yang
- Department of Anorectal Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
19
|
Yeh TH, Huang SY, Lan WY, Liaw GJ, Yu JY. Modulation of cell morphogenesis by tousled-like kinase in the Drosophila follicle cell. Dev Dyn 2015; 244:852-65. [PMID: 25981356 DOI: 10.1002/dvdy.24292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/30/2015] [Accepted: 05/07/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tousled-like kinase (Tlk) is a conserved serine/threonine kinase regulating DNA replication, chromatin assembly, and DNA repair. Previous studies have suggested that Tlk is involved in cell morphogenesis in vitro. In addition, tlk genetically interact with Rho1, which encodes a key regulator of the cytoskeleton. However, whether Tlk plays a physiological role in cell morphogenesis and cytoskeleton rearrangement remains unknown. RESULTS In tlk mutant follicle cells, area of the apical domain was reduced. The density of microtubules was increased in tlk mutant cells. The density of actin filaments was increased in the apical region and decreased in the basal region. Because area of the apical domain was reduced, we examined the levels of proteins located in the apical region by using immunofluorescence. The fluorescence intensities of two adherens junction proteins Armadillo (Arm) and DE-cadherin (DE-cad), atypical protein kinase C (aPKC), and Notch, were all increased in tlk mutant cells. The basolateral localized Discs large (Dlg) shifted apically in tlk mutant cells. CONCLUSIONS Increase of protein densities in the apical region might be resulted from disruption of the cytoskeleton and shrinkage of the apical domain. Together, these data suggest a novel role of Tlk in maintaining cell morphology, possibly through modulating the cytoskeleton.
Collapse
Affiliation(s)
- Tsung-Han Yeh
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Yu Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Yu Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Gwo-Jen Liaw
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
20
|
Mugat B, Akkouche A, Serrano V, Armenise C, Li B, Brun C, Fulga TA, Van Vactor D, Pélisson A, Chambeyron S. MicroRNA-Dependent Transcriptional Silencing of Transposable Elements in Drosophila Follicle Cells. PLoS Genet 2015; 11:e1005194. [PMID: 25993106 PMCID: PMC4451950 DOI: 10.1371/journal.pgen.1005194] [Citation(s) in RCA: 17] [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: 12/04/2014] [Accepted: 04/02/2015] [Indexed: 12/21/2022] Open
Abstract
RNA interference-related silencing mechanisms concern very diverse and distinct biological processes, from gene regulation (via the microRNA pathway) to defense against molecular parasites (through the small interfering RNA and the Piwi-interacting RNA pathways). Small non-coding RNAs serve as specificity factors that guide effector proteins to ribonucleic acid targets via base-pairing interactions, to achieve transcriptional or post-transcriptional regulation. Because of the small sequence complementarity required for microRNA-dependent post-transcriptional regulation, thousands of microRNA (miRNA) putative targets have been annotated in Drosophila. In Drosophila somatic ovarian cells, genomic parasites, such as transposable elements (TEs), are transcriptionally repressed by chromatin changes induced by Piwi-interacting RNAs (piRNAs) that prevent them from invading the germinal genome. Here we show, for the first time, that a functional miRNA pathway is required for the piRNA-mediated transcriptional silencing of TEs in this tissue. Global miRNA depletion, caused by tissue- and stage-specific knock down of drosha (involved in miRNA biogenesis), AGO1 or gawky (both responsible for miRNA activity), resulted in loss of TE-derived piRNAs and chromatin-mediated transcriptional de-silencing of TEs. This specific TE de-repression was also observed upon individual titration (by expression of the complementary miRNA sponge) of two miRNAs (miR-14 and miR-34) as well as in a miR-14 loss-of-function mutant background. Interestingly, the miRNA defects differentially affected TE- and 3' UTR-derived piRNAs. To our knowledge, this is the first indication of possible differences in the biogenesis or stability of TE- and 3' UTR-derived piRNAs. This work is one of the examples of detectable phenotypes caused by loss of individual miRNAs in Drosophila and the first genetic evidence that miRNAs have a role in the maintenance of genome stability via piRNA-mediated TE repression.
Collapse
Affiliation(s)
- Bruno Mugat
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Abdou Akkouche
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Vincent Serrano
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Claudia Armenise
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Blaise Li
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Christine Brun
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Tudor A. Fulga
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alain Pélisson
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| | - Séverine Chambeyron
- Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Montpellier, France
| |
Collapse
|
21
|
Du X, Wang H, Xu F, Huang Y, Liu Z, Liu T. Enterovirus 71 induces apoptosis of SH‑SY5Y human neuroblastoma cells through stimulation of endogenous microRNA let-7b expression. Mol Med Rep 2015; 12:953-9. [PMID: 25779425 PMCID: PMC4438926 DOI: 10.3892/mmr.2015.3482] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 07/21/2014] [Indexed: 11/05/2022] Open
Abstract
Enterovirus 71 (EV71) is a pathogenic microorganism that causes hand, foot and mouth disease. However, the epigenetic mechanisms behind how EV71 regulates host cell proliferation and apoptosis are unclear. In the present study, the ability of EV71 to induce apoptosis was analyzed in the SH-SY5Y human neuroblastoma cell line and the effect of this virus on the mRNA expression levels of various apoptotic markers, miRNA let-7b and cyclin D1 (CCND1), was also investigated. The results demonstrated that EV71 induced SH-SY5Y cell apoptosis. An MTT assay revealed a significant inhibitory effect of EV71 on cell proliferation between 12-72 h post injection, compared with the control group. Furthermore, quantitative polymerase chain reaction and western blot analyses demonstrated that expression level of the apoptosis inhibitor Bcl-2 was markedly reduced, but the expression levels of the apoptosis-promoting factors Bax, caspase-7, caspase‑3 and active caspase-3 were markedly higher in the SH-SY5Y cells 12-48 h after EV71 infection, compared with the non-infected cells. In addition, flow cytometric assays revealed that EV71 arrested the cell cycle of host SH-SY5Y cells. Northern blot analysis revealed a marked miRNA let-7b hybridization signal in the EV71 virus-infected group compared with the non-infected group. Furthermore, western blotting confirmed that the CCND1 protein expression levels were significantly reduced in EV71-infected SH-SY5Y cells. EV71-inhibited SH-SY5Y proliferation was abrogated using let-7b specific 2'-O-Methyl-RNA, which inhibited endogenous miRNA let-7b expression. Thus, EV71 regulated the host SH‑SY5Y cell cycle and cell proliferation via stimulating endo-genous miRNA let-7b and directly targeting CCND1, therefore EV71 is a potential candidate for antiviral therapy.
Collapse
Affiliation(s)
- Xiling Du
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Haipeng Wang
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Fuhui Xu
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Yongyi Huang
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Zhixue Liu
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Te Liu
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| |
Collapse
|
22
|
Abstract
In eukaryotic organisms, the orthologs of the DEAD-box RNA helicase Ded1p from yeast and DDX3 from human form a well-defined subfamily that is characterized by high sequence conservation in their helicase core and their N- and C- termini. Individual members of this Ded1/DDX3 subfamily perform multiple functions in RNA metabolism in both nucleus and cytoplasm. Ded1/DDX3 subfamily members have also been implicated in cellular signaling pathways and are targeted by diverse viruses. In this review, we discuss the considerable body of work on the biochemistry and biology of these proteins, including the recently discovered link of human DDX3 to tumorigenesis.
Collapse
Affiliation(s)
- Deepak Sharma
- Center for RNA Molecular Biology & Department of Biochemistry, School of Medicine, Case Western Reserve University , Cleveland, OH , USA
| | | |
Collapse
|
23
|
Olena AF, Rao MB, Thatcher EJ, Wu SY, Patton JG. miR-216a regulates snx5, a novel notch signaling pathway component, during zebrafish retinal development. Dev Biol 2015; 400:72-81. [PMID: 25645681 DOI: 10.1016/j.ydbio.2015.01.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 01/14/2015] [Accepted: 01/17/2015] [Indexed: 01/13/2023]
Abstract
Precise regulation of Notch signaling is essential for normal vertebrate development. Mind bomb (Mib) is a ubiquitin ligase that is required for activation of Notch by Notch׳s ligand, Delta. Sorting Nexin 5 (SNX5) co-localizes with Mib and Delta complexes and has been shown to directly bind to Mib. We show that microRNA-216a (miR-216a) is expressed in the retina during early development and regulates snx5 to precisely regulate Notch signaling. miR-216a and snx5 have complementary expression patterns. Knocking down miR-216a and/or overexpression of snx5 resulted in increased Notch activation. Conversely, knocking down snx5 and/or miR-216a overexpression caused a decrease in Notch activation. We propose a model in which SNX5, precisely controlled by miR-216a, is a vital partner of Mib in promoting endocytosis of Delta and subsequent activation of Notch signaling.
Collapse
Affiliation(s)
- Abigail F Olena
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| | - Mahesh B Rao
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| | | | - Shu-Yu Wu
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| |
Collapse
|
24
|
Palmer WH, Jia D, Deng WM. Cis-interactions between Notch and its ligands block ligand-independent Notch activity. eLife 2014; 3. [PMID: 25486593 PMCID: PMC4286723 DOI: 10.7554/elife.04415] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/06/2014] [Indexed: 01/01/2023] Open
Abstract
The Notch pathway is integrated into numerous developmental processes and therefore is fine-tuned on many levels, including receptor production, endocytosis, and degradation. Notch is further characterized by a twofold relationship with its Delta-Serrate (DSL) ligands, as ligands from opposing cells (trans-ligands) activate Notch, whereas ligands expressed in the same cell (cis-ligands) inhibit signaling. We show that cells without both cis- and trans-ligands can mediate Notch-dependent developmental events during Drosophila oogenesis, indicating ligand-independent Notch activity occurs when the receptor is free of cis- and trans-ligands. Furthermore, cis-ligands can reduce Notch activity in endogenous and genetically induced situations of elevated trans-ligand-independent Notch signaling. We conclude that cis-expressed ligands exert their repressive effect on Notch signaling in cases of trans-ligand-independent activation, and propose a new function of cis-inhibition which buffers cells against accidental Notch activity. DOI:http://dx.doi.org/10.7554/eLife.04415.001 Many biological processes require cells to send messages to one another. Typically, this is achieved when molecules are released from one cell and make contact with companion molecules on another cell. This triggers a chemical or biological reaction in the receiving cell. One of the most common examples of this is the Notch pathway, which is used throughout the animal kingdom and plays an important role in helping cells and embryos to develop. The Notch protein itself is a ‘receptor’ protein that is embedded in the surface of a cell, and relays signals from outside the cell to activate certain genes inside the cell. In fruit flies, two proteins called Serrate and Delta act as ‘ligands’ for Notch—by binding to Notch, they can change how this receptor works. If Serrate or Delta are present on the outside of one cell, they can activate Notch (and hence the Notch signaling pathway) in an adjacent cell. However, if the Serrate or Delta ligands are present on the surface of the same cell as Notch they turn the receptor off, rather than activate it. Notch can also work without being activated by Serrate or Delta, but whether the ligands can inhibit this ‘ligand-independent’ Notch activation if they are on the surface of the same cell as the Notch receptor was unknown. Palmer et al. study Notch signaling in the fruit fly equivalent of the ovary, in cells that are naturally deficient in Serrate and from which Delta was artificially removed. The Notch protein was activated when these ligands were not present. Furthermore, the developmental processes that are activated by Notch were able to proceed as normal when triggered by ligand-independent Notch signaling. In total, Palmer et al. investigated three different types of fruit fly cell, and found that ligand-independent Notch signaling can occur in all of them. Reintroducing Delta to the same cell as Notch turns the receptor off, suggesting that ligands on the surface of the same cell as the receptor can inhibit ligand-independent Notch activity. Many genetic diseases and cancers have been linked to Notch being activated when it should not be; therefore, understanding how Notch is controlled could help guide the development of new treatments for these conditions. DOI:http://dx.doi.org/10.7554/eLife.04415.002
Collapse
Affiliation(s)
- William Hunt Palmer
- Department of Biological Science, Florida State University, Tallahassee, United States
| | - Dongyu Jia
- Department of Biological Science, Florida State University, Tallahassee, United States
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, United States
| |
Collapse
|
25
|
Xie G, Yu Z, Jia D, Jiao R, Deng WM. E(y)1/TAF9 mediates the transcriptional output of Notch signaling in Drosophila. J Cell Sci 2014; 127:3830-9. [PMID: 25015288 DOI: 10.1242/jcs.154583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transcriptional activation of Notch signaling targets requires the formation of a ternary complex that involves the intracellular domain of the Notch receptor (NICD), DNA-binding protein Suppressor of Hairless [Su(H), RPBJ in mammals] and coactivator Mastermind (Mam). Here, we report that E(y)1/TAF9, a component of the transcription factor TFIID complex, interacts specifically with the NICD-Su(H)-Mam complex to facilitate the transcriptional output of Notch signaling. We identified E(y)1/TAF9 in a large-scale in vivo RNA interference (RNAi) screen for genes that are involved in a Notch-dependent mitotic-to-endocycle transition in Drosophila follicle cells. Knockdown of e(y)1/TAF9 displayed Notch-mutant-like phenotypes and defects in target gene and activity reporter expression in both the follicle cells and wing imaginal discs. Epistatic analyses in these two tissues indicated that E(y)1/TAF9 functions downstream of Notch cleavage. Biochemical studies in S2 cells demonstrated that E(y)1/TAF9 physically interacts with the transcriptional effectors of Notch signaling Su(H) and NICD. Taken together, our data suggest that the association of the NICD-Su(H)-Mastermind complex with E(y)1/TAF9 in response to Notch activation recruits the transcription initiation complex to induce Notch target genes, coupling Notch signaling with the transcription machinery.
Collapse
Affiliation(s)
- Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Zhongsheng Yu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China
| | - Dongyu Jia
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Renjie Jiao
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| |
Collapse
|
26
|
Sun F, Wan M, Xu X, Gao B, Zhou Y, Sun J, Cheng L, Klein OD, Zhou X, Zheng L. Crosstalk between miR-34a and Notch Signaling Promotes Differentiation in Apical Papilla Stem Cells (SCAPs). J Dent Res 2014; 93:589-95. [PMID: 24710391 DOI: 10.1177/0022034514531146] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 03/19/2014] [Indexed: 02/05/2023] Open
Abstract
Stem cells from the apical papilla (SCAPs) are important for the formation and regeneration of root dentin. Here, we examined the expression of Notch signaling components in SCAPs and investigated crosstalk between microRNA miR-34aand Notch signaling during cell differentiation. We found that human SCAPs express NOTCH2, NOTCH3, JAG2, DLL3, and HES1, and we tested the relationship between Notch signaling and both cell differentiation and miR-34a expression. NOTCH activation in SCAPs inhibited cell differentiation and up-regulated the expression of miR-34a, whereas miR-34a inhibited Notch signaling in SCAPs by directly targeting the 3'UTR of NOTCH2 and HES1 mRNA and suppressing the expression of NOTCH2, N2ICD, and HES1. DSPP, RUNX2, OSX, and OCN expression was consequently up-regulated. Thus, Notch signaling in human SCAPs plays a vital role in maintenance of these cells. miR-34a interacts with Notch signaling and promotes both odontogenic and osteogenic differentiation of SCAPs.
Collapse
Affiliation(s)
- F Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - M Wan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 Program in Craniofacial and Mesenchymal Biology and Departments of Orofacial Sciences and Pediatrics, University of California, San Francisco, CA 94143, USA
| | - X Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - B Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Y Zhou
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - J Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - L Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - O D Klein
- Program in Craniofacial and Mesenchymal Biology and Departments of Orofacial Sciences and Pediatrics, University of California, San Francisco, CA 94143, USA
| | - X Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| | - L Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041 West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China, 610041
| |
Collapse
|
27
|
Regulation of broad by the Notch pathway affects timing of follicle cell development. Dev Biol 2014; 392:52-61. [PMID: 24815210 DOI: 10.1016/j.ydbio.2014.04.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/23/2014] [Accepted: 04/29/2014] [Indexed: 12/20/2022]
Abstract
During Drosophila oogenesis, activation of Notch signaling in the follicular epithelium (FE) around stage 6 of oogenesis is essential for entry into the endocycle and a series of other changes such as cell differentiation and migration of subsets of the follicle cells. Notch induces the expression of zinc finger protein Hindsight and suppresses homeodomain protein Cut to regulate the mitotic/endocycle (ME) switch. Here we report that broad (br), encoding a small group of zinc-finger transcription factors resulting from alternative splicing, is a transcriptional target of Notch nuclear effector Suppressor of Hairless (Su(H)). The early pattern of Br in the FE, uniformly expressed except in the polar cells, is established by Notch signaling around stage 6, through the binding of Su(H) to the br early enhancer (brE) region. Mutation of the Su(H) binding site leads to a significant reduction of brE reporter expression in follicle cells undergoing the endocycle. Chromatin immunoprecipitation results further confirm Su(H) binding to the br early enhancer. Consistent with its expression in follicle cells during midoogenesis, loss of br function results in a delayed entry into the endocycle. Our findings suggest an important role of br in the timing of follicle cell development, and its transcriptional regulation by the Notch pathway.
Collapse
|
28
|
Berns N, Woichansky I, Friedrichsen S, Kraft N, Riechmann V. A genome-scale in vivo RNAi analysis of epithelial development in Drosophila identifies new proliferation domains outside of the stem cell niche. J Cell Sci 2014; 127:2736-48. [PMID: 24762813 DOI: 10.1242/jcs.144519] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Drosophila oogenesis system provides an excellent model to study the development of epithelial tissues. Here, we report the first genome-scale in vivo RNA interference (RNAi) screen for genes controlling epithelial development. By directly analysing cell and tissue architecture we identified 1125 genes, which we assigned to seven different functions in epithelial formation and homeostasis. We validated the significance of our screen by generating mutants for Vps60, a component of the endosomal sorting complexes required for transport (ESCRT) machinery. This analysis provided new insights into spatiotemporal control of cell proliferation in the follicular epithelium. Previous studies have identified signals controlling divisions in the follicle stem cell niche. However, 99% of cell divisions occur outside of the niche and it is unclear how these divisions are controlled. Our data distinguish two new domains outside of the stem cell niche where there are differing controls on proliferation. One domain abuts the niche and is characterised by ESCRT, Notch and JAK/STAT-mediated control of proliferation. Adjacent to this domain, another domain is defined by loss of the impact of ESCRT on cell division. Thus, during development epithelial cells pass through a variety of microenvironments that exert different modes of proliferation control. The switch between these modes might reflect a decrease in the 'stemness' of epithelial cells over time.
Collapse
Affiliation(s)
- Nicola Berns
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Innokenty Woichansky
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Steffen Friedrichsen
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Nadine Kraft
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Veit Riechmann
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
29
|
|
30
|
Asgari S. MicroRNA functions in insects. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2013; 43:388-97. [PMID: 23103375 DOI: 10.1016/j.ibmb.2012.10.005] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 10/11/2012] [Accepted: 10/16/2012] [Indexed: 05/14/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are generated in all eukaryotes and viruses. Their role as master regulators of gene expression in various biological processes has only been fully appreciated over the last decade. Accumulating evidence suggests that alterations in the expression of miRNAs may lead to disorders, including developmental defects, diseases and cancer. Here, I review what is currently known about miRNA functions in insects to provide an insight into their diverse roles in insect biology.
Collapse
Affiliation(s)
- Sassan Asgari
- School of Biological Sciences, The University of Queensland, Brisbane, St Lucia, QLD 4072, Australia.
| |
Collapse
|
31
|
Huang YC, Smith L, Poulton J, Deng WM. The microRNA miR-7 regulates Tramtrack69 in a developmental switch in Drosophila follicle cells. Development 2013; 140:897-905. [PMID: 23325762 DOI: 10.1242/dev.080192] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Development in multicellular organisms includes both small incremental changes and major switches of cell differentiation and proliferation status. During Drosophila oogenesis, the follicular epithelial cells undergo two major developmental switches that cause global changes in the cell-cycle program. One, the switch from the endoreplication cycle to a gene-amplification phase, during which special genomic regions undergo repeated site-specific replication, is attributed to Notch downregulation, ecdysone signaling activation and upregulation of the zinc-finger protein Tramtrack69 (Ttk69). Here, we report that the microRNA miR-7 exerts an additional layer of regulation in this developmental switch by regulating Ttk69 transcripts. miR-7 recognizes the 3' UTR of ttk69 transcripts and regulates Ttk69 expression in a dose-dependent manner. Overexpression of miR-7 effectively blocks the switch from the endocycle to gene amplification through its regulation of ttk69. miR-7 and Ttk69 also coordinate other cell differentiation events, such as vitelline membrane protein expression, that lead to the formation of the mature egg. Our studies reveal the important role miR-7 plays in developmental decision-making in association with signal-transduction pathways.
Collapse
Affiliation(s)
- Yi-Chun Huang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, USA
| | | | | | | |
Collapse
|
32
|
Abstract
One of the most important and evolutionarily conserved strategies to control gene expression in higher metazoa is posttranscriptional regulation via small regulatory RNAs such as microRNAs (miRNAs), endogenous small interfering RNAs (endo-siRNAs), and piwi-interacting RNAs (piRNAs). Primordial germ cells, which are defined by their totipotent potential and noted for their dependence on posttranscriptional regulation by RNA-binding proteins, rely on these small regulatory RNAs for virtually every aspect of their development, including specification, migration, and differentiation into competent gametes. Here, we review current knowledge of the roles miRNAs, endo-siRNAs, and piRNAs play at all stages of germline development in various organisms, focusing on studies in the mouse.
Collapse
Affiliation(s)
- Matthew S Cook
- Department of Urology, University of California, San Francisco, California, USA.
| | | |
Collapse
|
33
|
Song J, Kim D, Chun CH, Jin EJ. MicroRNA-375, a new regulator of cadherin-7, suppresses the migration of chondrogenic progenitors. Cell Signal 2012. [PMID: 23178988 DOI: 10.1016/j.cellsig.2012.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Endochondral bone formation requires a complex interplay among immature mesenchymal progenitor cells to form the cartilaginous anlagen, involving migration, aggregation and condensation. Even though condensation of chondrogenic progenitors is an essential step in this process, the mechanism(s) by which this occurs has not been well studied. Here, we investigated the involvement of microRNAs (miRNAs) in this process and found that the expression of miR-375 decreased upon chondrogenic differentiation of limb mesenchymal cells. Blockade of miR-375 via peptide nucleic acid (PNA)-based antisense oligonucleotides (ASOs) increased the migration of chondrogenic progenitors, the formation of precartilage condensations and the expression level of cadherin-7. Furthermore, miR-375 was necessary and sufficient to down-regulate cell migration through negative regulation of cadherin-7 by the direct interaction with 3' UTR of cadherin-7. In addition, miR-375 is also involved in the cell migration and precartilage condensation mediated by p38MAPK, a positive signaling in the chondrogenic differentiation. Collectively, our results suggest that miR-375 negatively modulates cell migration and subsequent precartilage condensation by targeting cadherin-7.
Collapse
Affiliation(s)
- Jinsoo Song
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, Republic of Korea
| | | | | | | |
Collapse
|
34
|
Efficient EGFR signaling and dorsal-ventral axis patterning requires syntaxin dependent Gurken trafficking. Dev Biol 2012; 373:349-58. [PMID: 23127433 DOI: 10.1016/j.ydbio.2012.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 11/22/2022]
Abstract
Vesicle trafficking plays a crucial role in the establishment of cell polarity in various cellular contexts, including axis-pattern formation in the developing egg chamber of Drosophila. The EGFR ligand, Gurken (Grk), is first localized at the posterior of young oocytes for anterior-posterior axis formation and later in the dorsal anterior region for induction of the dorsal-ventral (DV) axis, but regulation of Grk localization by membrane trafficking in the oocyte remains poorly understood. Here, we report that Syntaxin 1A (Syx1A) is required for efficient trafficking of Grk protein for DV patterning. We show that Syx1A is associated with the Golgi membrane and is required for the transportation of Grk-containing vesicles along the microtubules to their dorsal anterior destination in the oocyte. Our studies reveal that the Syx1A dependent trafficking of Grk protein is required for efficient EGFR signaling during DV patterning.
Collapse
|
35
|
Liu T, Cheng W, Gao Y, Wang H, Liu Z. Microarray analysis of microRNA expression patterns in the semen of infertile men with semen abnormalities. Mol Med Rep 2012; 6:535-42. [PMID: 22735917 DOI: 10.3892/mmr.2012.967] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 06/18/2012] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) play a crucial role in tissue development and the pathology of many diseases, however, the effects and roles of miRNAs in the development of semen abnormalities in infertile males have not yet been investigated. In this study, we analyzed and compared the miRNA expression profiles of abnormal semen from 86 infertile males with normal semen from 86 healthy males using an miRNA microarray. In total, 52 miRNAs were differentially expressed between the abnormal semen of infertile males and the normal semen of healthy males. The differential expression of selected miRNAs was validated by real time qRT-PCR and northern blotting: miR-574-5p, miR-297, miR-122, miR-1275, miR-373, miR-185 and miR-193b were upregulated (fold change>1.5, p<0.001) and miR-100, miR-512-3p, miR-16, miR-19b, miR-23b and miR-26a were downregulated (fold change<0.667, p<0.001) in the semen of infertile males with semen abnormalities. In conclusion, this study provides new insights into specific miRNAs that are associated with semen abnormalities in infertile males.
Collapse
Affiliation(s)
- Te Liu
- School of Environmental Science and Engineering, Donghua University, and International Peace Maternity and Child Health Hospital, Shanghai 201620, PR China.
| | | | | | | | | |
Collapse
|
36
|
Tanaka ED, Piulachs MD. Dicer-1 is a key enzyme in the regulation of oogenesis in panoistic ovaries. Biol Cell 2012; 104:452-61. [PMID: 22462497 DOI: 10.1111/boc.201100044] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 03/26/2012] [Indexed: 11/26/2022]
Abstract
BACKGROUND INFORMATION In insects, the action of microRNAs (miRNAs) on oogenesis has been explored only in dipterans, which possess meroistic ovaries, a highly modified ovarian type. Here we study miRNA function in the most primitive, panoistic type of ovaries using the phylogenetically basal insect Blattella germanica (Dictyoptera, Blattellidae) as model. RESULTS Dicer-1 (Dcr1), a key enzyme in miRNA biogenesis, was depleted using RNAi. Females treated with double-stranded RNA targeting Dicer-1, exhibited deep alterations in oocyte development; among them, the follicular epithelia of the basal oocytes did not develop, thus resulting in sterile females. CONCLUSIONS These effects derived from the absence of Dicer-1 suggest that miRNAs are crucial for the regulation of oogenesis in panoistic ovaries, the most primitive insect ovarian type.
Collapse
Affiliation(s)
- Erica Donato Tanaka
- Institut de Biologia Evolutiva (Universitat Pompeu Fabra-Consejo Superior de Investigaciones Cientificas), 08003 Barcelona, Spain
| | | |
Collapse
|
37
|
|