1
|
Gartner A, Engebrecht J. DNA repair, recombination, and damage signaling. Genetics 2021; 220:6522877. [PMID: 35137093 PMCID: PMC9097270 DOI: 10.1093/genetics/iyab178] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 01/09/2023] Open
Abstract
DNA must be accurately copied and propagated from one cell division to the next, and from one generation to the next. To ensure the faithful transmission of the genome, a plethora of distinct as well as overlapping DNA repair and recombination pathways have evolved. These pathways repair a large variety of lesions, including alterations to single nucleotides and DNA single and double-strand breaks, that are generated as a consequence of normal cellular function or by external DNA damaging agents. In addition to the proteins that mediate DNA repair, checkpoint pathways have also evolved to monitor the genome and coordinate the action of various repair pathways. Checkpoints facilitate repair by mediating a transient cell cycle arrest, or through initiation of cell suicide if DNA damage has overwhelmed repair capacity. In this chapter, we describe the attributes of Caenorhabditis elegans that facilitate analyses of DNA repair, recombination, and checkpoint signaling in the context of a whole animal. We review the current knowledge of C. elegans DNA repair, recombination, and DNA damage response pathways, and their role during development, growth, and in the germ line. We also discuss how the analysis of mutational signatures in C. elegans is helping to inform cancer mutational signatures in humans.
Collapse
Affiliation(s)
- Anton Gartner
- Department for Biological Sciences, IBS Center for Genomic Integrity, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea,Corresponding author: (A.G.); (J.E.)
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA,Corresponding author: (A.G.); (J.E.)
| |
Collapse
|
2
|
The X-linked trichothiodystrophy-causing gene RNF113A links the spliceosome to cell survival upon DNA damage. Nat Commun 2020; 11:1270. [PMID: 32152280 PMCID: PMC7062854 DOI: 10.1038/s41467-020-15003-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/17/2020] [Indexed: 12/30/2022] Open
Abstract
Prolonged cell survival occurs through the expression of specific protein isoforms generated by alternate splicing of mRNA precursors in cancer cells. How alternate splicing regulates tumor development and resistance to targeted therapies in cancer remain poorly understood. Here we show that RNF113A, whose loss-of-function causes the X-linked trichothiodystrophy, is overexpressed in lung cancer and protects from Cisplatin-dependent cell death. RNF113A is a RNA-binding protein which regulates the splicing of multiple candidates involved in cell survival. RNF113A deficiency triggers cell death upon DNA damage through multiple mechanisms, including apoptosis via the destabilization of the prosurvival protein MCL-1, ferroptosis due to enhanced SAT1 expression, and increased production of ROS due to altered Noxa1 expression. RNF113A deficiency circumvents the resistance to Cisplatin and to BCL-2 inhibitors through the destabilization of MCL-1, which thus defines spliceosome inhibitors as a therapeutic approach to treat tumors showing acquired resistance to specific drugs due to MCL-1 stabilization. Alternate splicing of mRNA precursors has been linked to tumor development. Here the authors reveal a role of the E3 ligase RNF113A in spliceosome regulation affecting cell survival upon DNA damage.
Collapse
|
3
|
Mendelsohn BA, Beleford DT, Abu-El-Haija A, Alsaleh NS, Rahbeeni Z, Martin PM, Rego S, Huang A, Capodanno G, Shieh JT, Van Ziffle J, Risch N, Alkuraya FS, Slavotinek AM. A novel truncating variant in ring finger protein 113A (RNF113A) confirms the association of this gene with X-linked trichothiodystrophy. Am J Med Genet A 2019; 182:513-520. [PMID: 31880405 DOI: 10.1002/ajmg.a.61450] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 11/09/2022]
Abstract
We describe an 11-year old boy with severe global developmental delays, failure to thrive and growth retardation, refractory seizures with recurrent status epilepticus, hypogammaglobulinemia, hypergonadotropic hypogonadism, and duodenal strictures. He had facial and skin findings compatible with trichothiodystrophy, including sparse and brittle hair, thin eyebrows, and dry skin. Exome sequencing showed a hemizygous, truncating variant in RNF113A, c.903_910delGCAGACCA, predicting p.(Gln302fs*12), that was inherited from his mother. Although his clinical features overlap closely with features described in the two previously reported male first cousins with RNF113A loss of function mutations, the duodenal strictures seen in this patient have not been reported. Interestingly, the patient's mother had short stature and 100% skewed X-inactivation as seen in other obligate female carriers. A second male with developmental delays, microcephaly, seizures, ambiguous genitalia, and facial anomalies that included sparse and brittle hair, thin eyebrows and dry skin was recently reported to have c.897_898delTG, predicting p.(Cys299*) in RNF113A and we provide additional clinical details for this patient. This report further supports deleterious variants in RNF113A as a cause of a novel trichothiodystrophy syndrome.
Collapse
Affiliation(s)
- Bryce A Mendelsohn
- Division of Medical Genetics, University of California, San Francisco, San Francisco, California
| | - Daniah T Beleford
- Division of Medical Genetics, University of California, San Francisco, San Francisco, California
| | - Aya Abu-El-Haija
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts.,Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
| | - Norah S Alsaleh
- Division of Genetics and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Zuhair Rahbeeni
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Pierre-Marie Martin
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Shannon Rego
- Division of Medical Genetics, University of California, San Francisco, San Francisco, California
| | - Alyssa Huang
- Division of Pediatric Endocrinology, University of California, San Francisco, California
| | - Gina Capodanno
- Division of Pediatric Endocrinology, University of California, San Francisco, California
| | - Joseph T Shieh
- Division of Medical Genetics, University of California, San Francisco, San Francisco, California.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Jessica Van Ziffle
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Neil Risch
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Fowzan S Alkuraya
- Division of Genetics and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Anne M Slavotinek
- Division of Medical Genetics, University of California, San Francisco, San Francisco, California.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| |
Collapse
|
4
|
Tessarech M, Gorce M, Boussion F, Bault J, Triau S, Charif M, Khiaty S, Delorme B, Guichet A, Ziegler A, Bris C, Laquerrière A, Fallet‐Bianco C, Jacquette A, Salhi H, Héron D, Reynier P, Procaccio V, Bonneau D, Colin E. Second report of RING finger protein 113A (
RNF113A)
involvement in a Mendelian disorder. Am J Med Genet A 2019; 182:565-569. [DOI: 10.1002/ajmg.a.61384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/01/2019] [Accepted: 09/26/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Marine Tessarech
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
| | - Magali Gorce
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
| | - Françoise Boussion
- Department of Obstetrics and GynecologyAngers University Hospital Angers France
| | - Jean‐Philippe Bault
- Department of Gynecology and ObstetricsCHI Poissy‐Saint‐Germain Poissy France
- Department of Gynecology and ObstetricsCH Bicêtre Kremlin‐Bicêtre France
| | - Stéphane Triau
- Department of PathologyAngers University Hospital Angers France
| | - Majida Charif
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Salim Khiaty
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Benoit Delorme
- Department of RadiologyAngers University Hospital Angers France
| | - Agnès Guichet
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
| | - Alban Ziegler
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Céline Bris
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Annie Laquerrière
- Normandie Univ, UNIROUEN, INSERM U1245Normandy Centre for Genomic and Personalized Medicine Rouen France
- Department of PathologyRouen University Hospital Rouen France
| | | | | | - Houria Salhi
- Department of PathologyCochin Saint Vincent de Paul Hospital, APHP Paris France
| | - Delphine Héron
- Department of Medical GeneticsTrousseau Hospital, APHP Paris France
| | - Pascal Reynier
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Vincent Procaccio
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Dominique Bonneau
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| | - Estelle Colin
- Department of Biochemistry and GeneticsAngers University Hospital Angers France
- UMR CNRS 6015‐INSERM 1083 and PREMMIMitovasc Institute Angers France
| |
Collapse
|
5
|
Tuerxun X, Abudumijiti H, Fen GC, Hasimu A. The functional role of RNF113A in cervical carcinogenesis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3570-3582. [PMID: 31934207 PMCID: PMC6949850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 08/27/2019] [Indexed: 06/10/2023]
Abstract
RNF113A is thought to function as an E3 ligase, engaged in the regulation of the turnover and activity of many target proteins. However, the fuctional role of RNF113A in cervical cancer remains unclear. In this study, by performing an immunohistochemistry (IHC) assay, we found that the RNF113A protein was significantly up-regulated in cervical cancer cells, and a high RNF113A expression was associated with malignant phenotypes. To determine the role of RNF113A in cervical cancer aggressiveness, we performed a gain and loss of functional experiments in cervical cancer cells with cell transfection, wound healing, transwell migration, and flow cytometry analysis. The results showed that RNF113A promotes the proliferation and survival ability of cervical cancer cells, enhances migration and invasion, and inhibits the apoptosis of cervical cancer cells. By silencing RNF113A in CSCC cell lines, we observed an up-regulation of the P53 protein level, indicating that P53 may function as a target of the RNF113A E3 ligase, and RNF113A may inhibit tumor cell apoptosis by degrading the TP53 protein.
Collapse
Affiliation(s)
- Xiayida Tuerxun
- Department of Pathology, College of Basic Medical Sciences, Xinjiang Medical UniversityUrumqi, Xinjiang Uyghur Autonomous Region, China
| | - Huerxidan Abudumijiti
- Department of Pathology, College of Basic Medical Sciences, Xinjiang Medical UniversityUrumqi, Xinjiang Uyghur Autonomous Region, China
| | - Guo Chun Fen
- Department of Gynecology in The First Affiliated Hospital, Medical University of XinjiangUrumqi, China
| | - Axiangu Hasimu
- Department of Pathology, College of Basic Medical Sciences, Xinjiang Medical UniversityUrumqi, Xinjiang Uyghur Autonomous Region, China
| |
Collapse
|
6
|
Gatti da Silva GH, Jurica MS, Chagas da Cunha JP, Oliveira CC, Coltri PP. Human RNF113A participates of pre-mRNA splicing in vitro. J Cell Biochem 2019; 120:8764-8774. [PMID: 30506991 DOI: 10.1002/jcb.28163] [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] [Received: 03/02/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
Abstract
Pre-messenger RNA (mRNA) splicing is an essential step in the control of eukaryotic gene expression. During splicing, the introns are removed from the gene transcripts as the exons are ligated to create mature mRNA sequences. Splicing is performed by the spliceosome, which is a macromolecular complex composed of five small nuclear RNAs (snRNAs) and more than 100 proteins. Except for the core snRNP proteins, most spliceosome proteins are transiently associated and presumably involved with the regulation of spliceosome activity. In this study, we explored the association and participation of the human protein RNF113A in splicing. The addition of excess recombinant RNF113A to in vitro splicing reactions results in splicing inhibition. In whole-cell lysates, RNF113A co-immunoprecipitated with U2, U4, and U6 snRNAs, which are components of the tri-snRNP, and with proteins PRP19 and BRR2. When HeLa cells were CRISPR-edited to reduce the RNF113A levels, the in vitro splicing efficiency was severely affected. Consistently, the splicing activity was partially restored after the addition of the recombinant GST-RNF113A. On the basis on these results, we propose a model in which RNF113A associates with the spliceosome by interacting with PRP19, promoting essential rearrangements that lead to splicing.
Collapse
Affiliation(s)
- Guilherme H Gatti da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Melissa S Jurica
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, California
| | | | - Carla C Oliveira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Patricia P Coltri
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, California.,Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Wang L, Hou Z, Hasim A, Abuduerheman A, Zhang H, Niyaz M, Awut I, Upur H, Sheyhidin I. RNF113A promotes the proliferation, migration and invasion, and is associated with a poor prognosis of esophageal squamous cell carcinoma. Int J Oncol 2018; 52:861-871. [PMID: 29393393 DOI: 10.3892/ijo.2018.4253] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 12/04/2017] [Indexed: 11/06/2022] Open
Abstract
Ring finger protein 113A (RNF113A) possesses a C3HC4 zinc finger domain and this domain is found in E3 ubiquitin ligase and is involved in tumorigenesis. To date, and at least to the best of our knowledge, there are no studies available which have investigated RNF113A in cancer. Thus, this study aimed to explore the role of RNF113A in the development of esophageal squamous cell carcinoma (ESCC). For this purpose, paraffin-embedded samples from 117 patients with ESCC were selected, as well as 41 pairs of fresh-frozen ESCC and adjacent normal tissue samples. RNF113A expression was examined by immunohistochemistry and reverse transcription-quantitative PCR (RT-qPCR). RNF113A was overexpressed or silenced in the EC9706 and Eca109 cells. The cells were examined for cell cycle progression, apoptosis, invasiveness and migration. Xenograft tumors were also created in mice using the Eca109 cells. Tumor differentiation (P=0.008) and T classification (P<0.001) were found to be significantly associated with RNF113A expression. No statistically significant association was observed between RNF113A expression and sex, age, histological type, tumor location and lymph node metastasis (N classification). Kaplan-Meier analysis revealed that the patients with ESCC with ahigh expression of RNF113A had a lower survival rate than those with a low expression (P=0.002). Multivariate analysis revealed that RNF113A expression (HR=2.406; 95% CI, 1.301-4.449, P=0.005) was independently associated with overall survival in patients with ESCC. The overexpression of RNF113A promoted proliferation, migration, and invasiveness of ESCC cell lines in vitro, and RNF113A silencing reversed these malignant behaviors. RNF113A knockdown inhibited tumor growth in vivo. Thus, these results indicate that RNF113A promotes the proliferation, migration and invasiveness of ESCC cell lines. RNF113A expression in ESCC is this associated with a poor prognosis of affected patients.
Collapse
Affiliation(s)
- Lei Wang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Zhichao Hou
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ayshamgul Hasim
- Department of Pathology, Medical University of Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Abulajiang Abuduerheman
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Haiping Zhang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Madiniyat Niyaz
- Clinical Medical Research Institute, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Idiris Awut
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Halmurat Upur
- Department of Uyghur Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| |
Collapse
|
8
|
Shear force-based genetic screen reveals negative regulators of cell adhesion and protrusive activity. Proc Natl Acad Sci U S A 2017; 114:E7727-E7736. [PMID: 28847951 DOI: 10.1073/pnas.1616600114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The model organism Dictyostelium discoideum has greatly facilitated our understanding of the signal transduction and cytoskeletal pathways that govern cell motility. Cell-substrate adhesion is downstream of many migratory and chemotaxis signaling events. Dictyostelium cells lacking the tumor suppressor PTEN show strongly impaired migratory activity and adhere strongly to their substrates. We reasoned that other regulators of migration could be obtained through a screen for overly adhesive mutants. A screen of restriction enzyme-mediated integration mutagenized cells yielded numerous mutants with the desired phenotypes, and the insertion sites in 18 of the strains were mapped. These regulators of adhesion and motility mutants have increased adhesion and decreased motility. Characterization of seven strains demonstrated decreased directed migration, flatness, increased filamentous actin-based protrusions, and increased signal transduction network activity. Many of the genes share homology to human genes and demonstrate the diverse array of cellular networks that function in adhesion and migration.
Collapse
|
9
|
Caenorhabditis elegans as a powerful alternative model organism to promote research in genetic toxicology and biomedicine. Arch Toxicol 2017; 91:2029-2044. [PMID: 28299394 DOI: 10.1007/s00204-017-1944-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
Abstract
In view of increased life expectancy the risk for disturbed integrity of genetic information increases. This inevitably holds the implication for higher incidence of age-related diseases leading to considerable cost increase in health care systems. To develop preventive strategies it is crucial to evaluate external and internal noxae as possible threats to our DNA. Especially the interplay of DNA damage response (DDR) and DNA repair (DR) mechanisms needs further deciphering. Moreover, there is a distinct need for alternative in vivo test systems for basic research and also risk assessment in toxicology. Especially the evaluation of combinational toxicity of environmentally present genotoxins and adverse effects of clinically used DNA damaging anticancer drugs is a major challenge for modern toxicology. This review focuses on the applicability of Caenorhabditis elegans as a model organism to unravel and tackle scientific questions related to the biological consequences of genotoxin exposure and highlights methods for studying DDR and DR. In this regard large-scale in vivo screens of mixtures of chemicals and extensive parallel sequencing are highlighted as unique advantages of C. elegans. In addition, concise information regarding evolutionary conserved molecular mechanisms of the DDR and DR as well as currently available data obtained from the use of prototypical genotoxins and preferential read-outs of genotoxin testing are discussed. The use of established protocols, which are already available in the community, is encouraged to facilitate and further improve the implementation of C. elegans as a powerful genetic model system in genetic toxicology and biomedicine.
Collapse
|
10
|
Son M, Kawasaki I, Oh BK, Shim YH. LIN-23, an E3 Ubiquitin Ligase Component, Is Required for the Repression of CDC-25.2 Activity during Intestinal Development in Caenorhabditis elegans. Mol Cells 2016; 39:834-840. [PMID: 27871172 PMCID: PMC5125940 DOI: 10.14348/molcells.2016.0238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/02/2016] [Accepted: 11/07/2017] [Indexed: 11/27/2022] Open
Abstract
Caenorhabditis elegans (C. elegans) utilizes two different cell-cycle modes, binucleations during the L1 larval stage and endoreduplications at four larval moltings, for its postembryonic intestinal development. Previous genetic studies indicated that CDC-25.2 is specifically required for binucleations at the L1 larval stage and is repressed before endoreduplications. Furthermore, LIN-23, the C. elegans β-TrCP ortholog, appears to function as a repressor of CDC-25.2 to prevent excess intestinal divisions. We previously reported that intestinal hyperplasia in lin-23(e1883) mutants was effectively suppressed by the RNAi depletion of cdc-25.2. Nevertheless, LIN-23 targeting CDC-25.2 for ubiquitination as a component of E3 ubiquitin ligase has not yet been tested. In this study, LIN-23 is shown to be the major E3 ubiquitin ligase component, recognizing CDC-25.2 to repress their activities for proper transition of cell-cycle modes during the C. elegans postembryonic intestinal development. In addition, for the first time that LIN-23 physically interacts with both CDC-25.1 and CDC-25.2 and facilitates ubiquitination for timely regulation of their activities during the intestinal development.
Collapse
Affiliation(s)
- Miseol Son
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Ichiro Kawasaki
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Bong-Kyeong Oh
- Institute of Medical Science, Hanyang University College of Medicine, Seoul 04763,
Korea
| | - Yhong-Hee Shim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
11
|
Corbett MA, Dudding-Byth T, Crock PA, Botta E, Christie LM, Nardo T, Caligiuri G, Hobson L, Boyle J, Mansour A, Friend KL, Crawford J, Jackson G, Vandeleur L, Hackett A, Tarpey P, Stratton MR, Turner G, Gécz J, Field M. A novel X-linked trichothiodystrophy associated with a nonsense mutation in RNF113A. J Med Genet 2015; 52:269-74. [DOI: 10.1136/jmedgenet-2014-102418] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|