1
|
Xie P, Guan J, He X, Zhao Z, Guo Y, Sun Z, Yao L, Lee TY, Chiang YC. CAP-m7G: A capsule network-based framework for specific RNA N7-methylguanosine site identification using image encoding and reconstruction layers. Comput Struct Biotechnol J 2025; 27:804-812. [PMID: 40109445 PMCID: PMC11919597 DOI: 10.1016/j.csbj.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Abstract
N7-methylguanosine (m7G) modifications play a pivotal role in RNA stability, mRNA export, and protein translation. They are closely associated with ribosome function and the regulation of gene expression. Dysregulation of m7G has been implicated in various diseases, including cancers and neurodegenerative disorders, where the loss of m7G can lead to genomic instability and uncontrolled cell proliferation. Accurate identification of m7G sites is thus essential for elucidating these mechanisms. Due to the high cost of experimentally validating m7G sites, several artificial intelligence models have been developed to predict these sites. However, the performance of these models is not yet optimal, and a user-friendly web server is still needed. To address these issues, we developed CAP-m7G, an innovative model that integrates Chaos Game Representation, Capsule Networks, and reconstruction layers. CAP-m7G achieved an accuracy of 96.63%, a specificity of 95.07%, and a Matthews correlation coefficient (MCC) of 0.933 on independent test data. Our results demonstrate that the integration of Chaos Game Representation with Capsule Network can effectively capture the crucial sequence information associated with m7G sites. The web server can be accessed at https://awi.cuhk.edu.cn/~biosequence/CAP-m7G/index.php.
Collapse
Affiliation(s)
- Peilin Xie
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Jiahui Guan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Xuxin He
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Zhihao Zhao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Yilin Guo
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Zhenglong Sun
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Lantian Yao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ying-Chih Chiang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Blvd, Longgang District, 518172, Shenzhen, China
| |
Collapse
|
2
|
Rajab SAS, Andersen LK, Kenter LW, Berlinsky DL, Borski RJ, McGinty AS, Ashwell CM, Ferket PR, Daniels HV, Reading BJ. Combinatorial metabolomic and transcriptomic analysis of muscle growth in hybrid striped bass (female white bass Morone chrysops x male striped bass M. saxatilis). BMC Genomics 2024; 25:580. [PMID: 38858615 PMCID: PMC11165755 DOI: 10.1186/s12864-024-10325-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 04/19/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Understanding growth regulatory pathways is important in aquaculture, fisheries, and vertebrate physiology generally. Machine learning pattern recognition and sensitivity analysis were employed to examine metabolomic small molecule profiles and transcriptomic gene expression data generated from liver and white skeletal muscle of hybrid striped bass (white bass Morone chrysops x striped bass M. saxatilis) representative of the top and bottom 10 % by body size of a production cohort. RESULTS Larger fish (good-growth) had significantly greater weight, total length, hepatosomatic index, and specific growth rate compared to smaller fish (poor-growth) and also had significantly more muscle fibers of smaller diameter (≤ 20 µm diameter), indicating active hyperplasia. Differences in metabolomic pathways included enhanced energetics (glycolysis, citric acid cycle) and amino acid metabolism in good-growth fish, and enhanced stress, muscle inflammation (cortisol, eicosanoids) and dysfunctional liver cholesterol metabolism in poor-growth fish. The majority of gene transcripts identified as differentially expressed between groups were down-regulated in good-growth fish. Several molecules associated with important growth-regulatory pathways were up-regulated in muscle of fish that grew poorly: growth factors including agt and agtr2 (angiotensins), nicotinic acid (which stimulates growth hormone production), gadd45b, rgl1, zfp36, cebpb, and hmgb1; insulin-like growth factor signaling (igfbp1 and igf1); cytokine signaling (socs3, cxcr4); cell signaling (rgs13, rundc3a), and differentiation (rhou, mmp17, cd22, msi1); mitochondrial uncoupling proteins (ucp3, ucp2); and regulators of lipid metabolism (apoa1, ldlr). Growth factors pttg1, egfr, myc, notch1, and sirt1 were notably up-regulated in muscle of good-growing fish. CONCLUSION A combinatorial pathway analysis using metabolomic and transcriptomic data collectively suggested promotion of cell signaling, proliferation, and differentiation in muscle of good-growth fish, whereas muscle inflammation and apoptosis was observed in poor-growth fish, along with elevated cortisol (an anti-inflammatory hormone), perhaps related to muscle wasting, hypertrophy, and inferior growth. These findings provide important biomarkers and mechanisms by which growth is regulated in fishes and other vertebrates as well.
Collapse
Affiliation(s)
- Sarah A S Rajab
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Linnea K Andersen
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Linas W Kenter
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, NH, USA
| | - David L Berlinsky
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, NH, USA
| | - Russell J Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Andrew S McGinty
- North Carolina State University, Pamlico Aquaculture Field Laboratory, Aurora, NC, USA
| | - Christopher M Ashwell
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Peter R Ferket
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Harry V Daniels
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA
| | - Benjamin J Reading
- Department of Applied Ecology, North Carolina State University, 100 Eugene Brooks Avenue, Box 7617, Raleigh, NC, 27695, USA.
- North Carolina State University, Pamlico Aquaculture Field Laboratory, Aurora, NC, USA.
| |
Collapse
|
3
|
Butera G, Manfredi M, Fiore A, Brandi J, Pacchiana R, De Giorgis V, Barberis E, Vanella V, Galasso M, Scupoli MT, Marengo E, Cecconi D, Donadelli M. Tumor Suppressor Role of Wild-Type P53-Dependent Secretome and Its Proteomic Identification in PDAC. Biomolecules 2022; 12:305. [PMID: 35204804 PMCID: PMC8869417 DOI: 10.3390/biom12020305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/10/2022] Open
Abstract
The study of the cancer secretome is gaining even more importance in cancers such as pancreatic ductal adenocarcinoma (PDAC), whose lack of recognizable symptoms and early detection assays make this type of cancer highly lethal. The wild-type p53 protein, frequently mutated in PDAC, prevents tumorigenesis by regulating a plethora of signaling pathways. The importance of the p53 tumor suppressive activity is not only primarily involved within cells to limit tumor cell proliferation but also in the extracellular space. Thus, loss of p53 has a profound impact on the secretome composition of cancer cells and marks the transition to invasiveness. Here, we demonstrate the tumor suppressive role of wild-type p53 on cancer cell secretome, showing the anti-proliferative, apoptotic and chemosensitivity effects of wild-type p53 driven conditioned medium. By using high-resolution SWATH-MS technology, we characterized the secretomes of p53-deficient and p53-expressing PDAC cells. We found a great number of secreted proteins that have known roles in cancer-related processes, 30 of which showed enhanced and 17 reduced secretion in response to p53 silencing. These results are important to advance our understanding on the link between wt-p53 and cancer microenvironment. In conclusion, this approach may detect a secreted signature specifically driven by wild-type p53 in PDAC.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Virginia Vanella
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Marilisa Galasso
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Department of Medicine, Section of Hematology, University of Verona, 37134 Verona, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, 37134 Verona, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| |
Collapse
|
4
|
Li Q, Xiao M, Shi Y, Hu J, Bi T, Wang C, Yan L, Li X. eIF5B regulates the expression of PD-L1 in prostate cancer cells by interacting with Wig1. BMC Cancer 2021; 21:1022. [PMID: 34525951 PMCID: PMC8442339 DOI: 10.1186/s12885-021-08749-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/28/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Eukaryotic translation initiation factors (eIFs) are the key factors to synthesize translation initiation complexes during the synthesis of eukaryotic proteins. Besides, eIFs are especially important in regulating the immune function of tumor cells. However, the effect mechanism of eIFs in prostate cancer remains to be studied, which is precisely the purpose of this study. METHODS In this study, three groups of prostate cancer cells were investigated. One group had its eIF5B gene knocked down; another group had its Programmed death 1 (PD-L1) overexpressed; the final group had its Wild-type p53-induced gene 1 (Wig1) overexpressed. Genetic alterations of the cancer cells were performed by plasmid transfection. The expression of PD-L1 mRNA was detected by quantitative real-time PCR (qRT-PCR), and the expressions of PD-L1 and eIF5B proteins were observed by western blot assays. Cell Counting Kit-8 (CCK-8), flow cytometry, Transwell and Transwell martrigel were used to investigated cell proliferation, apoptosis, migration and invasion, respectively. The effect of peripheral blood mononuclear cells (PBMCs) on tumor cells was observed, and the interaction between eIF5B and Wig1 was revealed by co-immunoprecipitation (CoIP) assay. Finally, the effects of interference with eIF5B expression on the growth, morphology, and immunity of the tumor, as well as PD-L1 expression in the tumor, were verified by tumor xenograft assays in vivo. RESULTS Compared with normal prostate epithelial cells, prostate cancer cells revealed higher expressions of eIF5B and PD-L1 interference with eIF-5B expression can inhibit the proliferation, migration, invasion and PD-L1 expression of prostate cancer cells. Meanwhile, the cancer cell group with interference with eIF5B expression also demonstrated greater, apoptosis and higher vulnerability to PBMCs. CoIP assays showed that Wig1 could bind to eIF5B in prostate cancer cells, and its overexpression can inhibit the proliferation, migration, invasion and PD-L1 expression of cancer cells while promoting apoptosis. Moreover, interference with eIF5B expression can inhibit tumor growth, destroy tumor morphology, and suppress the proliferation of tumor cells. CONCLUSION eIF5B can promote the expression of PD-L1 by interacting with Wig1. Besides, interference with eIF5B expression can inhibit the proliferation, migration, invasion and immunosuppressive response of prostate cancer cells. This study proposes a new target, eIF5B, for immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Qi Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China.
| | - Mulun Xiao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Yibo Shi
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Jinhao Hu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Tianxiang Bi
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Chaoliang Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Liang Yan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, China
| | - Xiaoyan Li
- Department of Neonatel Intensive Care Unit, Zhengzhou First People's Hospital, Zhengzhou City, 450004, Henan Province, China
| |
Collapse
|
5
|
Lin KH, Wilson GM, Blanco R, Steinert ND, Zhu WG, Coon JJ, Hornberger TA. A deep analysis of the proteomic and phosphoproteomic alterations that occur in skeletal muscle after the onset of immobilization. J Physiol 2021; 599:2887-2906. [PMID: 33873245 PMCID: PMC8353513 DOI: 10.1113/jp281071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/06/2021] [Indexed: 02/04/2023] Open
Abstract
KEY POINTS A decrease in protein synthesis plays a major role in the loss of muscle mass that occurs in response to immobilization. In mice, immobilization leads to a rapid (within 6 h) and progressive decrease in the rate of protein synthesis and this effect is mediated by a decrease in translational efficiency. Deep proteomic and phosphoproteomic analyses of mouse skeletal muscles revealed that the rapid immobilization-induced decrease in protein synthesis cannot be explained by changes in the abundance or phosphorylation state of proteins that have been implicated in the regulation of translation. ABSTRACT The disuse of skeletal muscle, such as that which occurs during immobilization, can lead to the rapid loss of muscle mass, and a decrease in the rate of protein synthesis plays a major role in this process. Indeed, current dogma contends that the decrease in protein synthesis is mediated by changes in the activity of protein kinases (e.g. mTOR); however, the validity of this model has not been established. Therefore, to address this, we first subjected mice to 6, 24 or 72 h of unilateral immobilization and then used the SUnSET technique to measure changes in the relative rate of protein synthesis. The result of our initial experiments revealed that immobilization leads to a rapid (within 6 h) and progressive decrease in the rate of protein synthesis and that this effect is mediated by a decrease in translational efficiency. We then performed a deep mass spectrometry-based analysis to determine whether this effect could be explained by changes in the expression and/or phosphorylation state of proteins that regulate translation. From this analysis, we were able to quantify 4320 proteins and 15,020 unique phosphorylation sites, and surprisingly, the outcomes revealed that the rapid immobilization-induced decrease in protein synthesis could not be explained by changes in either the abundance, or phosphorylation state, of proteins. The results of our work not only challenge the current dogma in the field, but also provide an expansive resource of information for future studies that are aimed at defining how disuse leads to loss of muscle mass.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Gary M Wilson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Rocky Blanco
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nathaniel D Steinert
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Wenyuan G Zhu
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
6
|
Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and Challenges in the Delivery of mRNA-based Vaccines. Pharmaceutics 2020; 12:E102. [PMID: 32013049 PMCID: PMC7076378 DOI: 10.3390/pharmaceutics12020102] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 02/08/2023] Open
Abstract
In the past few years, there has been increasing focus on the use of messenger RNA (mRNA) as a new therapeutic modality. Current clinical efforts encompassing mRNA-based drugs are directed toward infectious disease vaccines, cancer immunotherapies, therapeutic protein replacement therapies, and treatment of genetic diseases. However, challenges that impede the successful translation of these molecules into drugs are that (i) mRNA is a very large molecule, (ii) it is intrinsically unstable and prone to degradation by nucleases, and (iii) it activates the immune system. Although some of these challenges have been partially solved by means of chemical modification of the mRNA, intracellular delivery of mRNA still represents a major hurdle. The clinical translation of mRNA-based therapeutics requires delivery technologies that can ensure stabilization of mRNA under physiological conditions. Here, we (i) review opportunities and challenges in the delivery of mRNA-based therapeutics with a focus on non-viral delivery systems, (ii) present the clinical status of mRNA vaccines, and (iii) highlight perspectives on the future of this promising new type of medicine.
Collapse
Affiliation(s)
| | | | | | | | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
7
|
Eukaryotic initiation factor 3, subunit C silencing inhibits cell proliferation and promotes apoptosis in human ovarian cancer cells. Biosci Rep 2019; 39:BSR20191124. [PMID: 31316002 PMCID: PMC6685053 DOI: 10.1042/bsr20191124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/02/2019] [Accepted: 07/14/2019] [Indexed: 01/23/2023] Open
Abstract
Ovarian cancer remains the leading cause of death among all gynaecological cancers, illustrating the urgent need to understand the molecular mechanisms involved in this disease. Eukaryotic initiation factor 3c (EIF3c) plays an important role in protein translation and cancer cell growth and proliferation, but its role in human ovarian cancer is unclear. Our results showed that EIF3c silencing significantly up-regulated 217 and down-regulated 340 genes. Ingenuity Pathway Analysis (IPA) indicated that the top differentially expressed genes are involved in ‘Classical Pathways’, ‘Diseases and Functions’ and ‘Networks’, especially those involved in signalling and cellular growth and proliferation. In addition, eIF3c silencing inhibited cellular proliferation, enhanced apoptosis and regulated the expression of apoptosis-associated proteins. In conclusion, these results indicate that by dysregulating translational initiation, eIF3c plays an important role in the proliferation and survival of human ovarian cancer cells. These results should provide experimental directions for further in-depth studies on important human ovarian cancer cell pathways.
Collapse
|
8
|
Impact of Heat Shock Protein 90 Inhibition on the Proteomic Profile of Lung Adenocarcinoma as Measured by Two-Dimensional Electrophoresis Coupled with Mass Spectrometry. Cells 2019; 8:cells8080806. [PMID: 31370342 PMCID: PMC6721529 DOI: 10.3390/cells8080806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/28/2019] [Accepted: 07/28/2019] [Indexed: 02/06/2023] Open
Abstract
Heat shock protein 90 (HSP90) is an important chaperone in lung adenocarcinoma, with relevant protein drivers such as EGFR (epidermal growth factor receptor) and EML4-ALK (echinoderm microtubule-associated protein-like protein4 fused to anaplastic lymphoma kinase) depending on it for their correct function, therefore HSP90 inhibitors show promise as potential treatments for lung adenocarcinoma. To study responses to its inhibition, HSP90 was pharmacologically interrupted by geldanamycin and resorcinol derivatives or with combined inhibition of HSP90 plus HSP70 in lung adenocarcinoma cell lines. Two-dimensional electrophoresis was performed to identify proteomic profiles associated with inhibition which will help to understand the biological basis for the responses. HSP90 inhibition resulted in altered protein profiles that differed according the treatment condition studied. Results revealed 254 differentially expressed proteins after treatments, among which, eukaryotic translation initiation factor3 subunit I (eIF3i) and citrate synthase demonstrated their potential role as response biomarkers. The differentially expressed proteins also enabled signalling pathways involved in responses to be identified; these included apoptosis, serine-glycine biosynthesis and tricarboxylic acid cycle. The proteomic profiles identified here contribute to an improved understanding of HSP90 inhibition and open possibilities for the detection of potential response biomarkers which will be essential to maximize treatment efficacy in lung adenocarcinoma.
Collapse
|
9
|
Pan Y, Zhang ZN, Yin LB, Fu YJ, Jiang YJ, Shang H. Reduced eIF3d accelerates HIV disease progression by attenuating CD8+ T cell function. J Transl Med 2019; 17:167. [PMID: 31118081 PMCID: PMC6530059 DOI: 10.1186/s12967-019-1925-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/15/2019] [Indexed: 01/22/2023] Open
Abstract
Background In human immunodeficiency virus (HIV) infection, 10–15% of individuals exhibit a rapid decline in CD4+ T cells and become rapid progressors (RPs). Overall, understanding the factors affecting rapid disease progression in early HIV infection (EHI) can aid in treatment initiation. Recent studies show that eIF3s, classic scaffold proteins during the translation initiation process, can directly promote or inhibit the translation of mRNA, therefore participating in the regulation of cell function. However, to our knowledge, it has not been addressed whether eIF3s are involved in the diverse prognosis of HIV infection. Methods Expression of eIF3s in primary cells from early or chronic HIV-infected patients was detected by real-time PCR. To investigate the potential mechanisms of eIF3d in the regulation of CD8+ T cell function, complete transcriptomes of eIF3d-inhibited Jurkat T cells were sequenced by RNA sequencing (RNA-Seq). Additionally, to examine the effect of eIF3d on CD8+ T cell function, eIF3d expression was inhibited alone or in combination with SOCS-7 knockdown by siRNA in isolated CD8+ T cells. CD8+ T cell proliferation, IFN-r secretion and apoptosis were detected by flow cytometry. Moreover, the effect of eIF3d on HIV replication was evaluated in Jurkat cells, peripheral blood mononuclear cells (PBMCs) and CD4+ T cells with eIF3d knockdown using a pNL4-3 pseudotyped virus. Results At approximately 100 days of infection, only eIF3d was markedly decreased in RPs compared with chronic progressors (CPs). Expression of eIF3d correlated significantly with disease progression in EHI. Based on in vitro analyses, reduced eIF3d expression led to decreased proliferation and IFN-γ secretion and increased apoptosis in CD8+ T cells. Inhibited expression of eIF3d caused enhanced expression of SOCS-7, and inhibiting SOCS-7 expression by siRNA rescued the attenuated CD8+ T cell function caused by eIF3d. Finally, when eIF3d was inhibited in Jurkat cells, PBMCs and CD4+ T cells, pNL4-3-VSV-G virus replication was enhanced. Conclusions The current data highlight the importance of eIF3d in HIV infection by inhibiting CD8+ T cell function and promoting viral replication. Our study provides potential targets for improved immune intervention. Electronic supplementary material The online version of this article (10.1186/s12967-019-1925-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ying Pan
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China
| | - Zi-Ning Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China
| | - Lin-Bo Yin
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China
| | - Ya-Jing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China
| | - Yong-Jun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, He ping District, Shenyang, Liaoning, 110001, China. .,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China. .,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 79 Qing Chun Street, Hangzhou, 310003, China.
| |
Collapse
|
10
|
Gao W, Hu Y, Zhang Z, Du G, Yin L, Yin Z. Knockdown of EIF3C promotes human U-2OS cells apoptosis through increased CASP3/7 and Chk1/2 by upregulating SAPK/JNK. Onco Targets Ther 2019; 12:1225-1235. [PMID: 30863090 PMCID: PMC6389005 DOI: 10.2147/ott.s187209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background As a component of the EIF3 complex, EIF3C is essential for several steps in protein synthesis initiation. Recently, it has been addressed that EIF3C is overexpressed in several human cancers and plays a pivotal role in cell proliferation and tumorigenesis. Materials and methods Immunohistochemistry, quantitative real-time PCR (qPCR), and Western blotting assays were employed to determine the expression of EIF3C in osteosarcoma (OsC) tissues obtained from 60 patients. The levels of EIF3C mRNA and protein were assessed by qPCR and Western blotting, respectively. The effect of EIF3C knockdown on OsC cell proliferation was detected by MTT and colony formation assays, respectively. Cell apoptosis induced by EIF3C silencing was analyzed by flow cytometric analysis. PathScan stress and apoptosis signaling antibody array kit was used to analyze the potential effects of EIF3C knockdown on OsC cells. Results The levels of EIF3C were high in OsC tissues and cell lines. In addition, EIF3C knockdown by lentivirus-mediated shRNA targeting EIF3C significantly suppressed cell proliferation and colony formation and induced apoptosis in U-2OS cells. Moreover, EIF3C knockdown led to the upregulated expression of CASP3/7, Chk1/2, and SAPK/JNK, indicating that the downregulated expression of EIF3C might be associated with pro-apoptosis of U-2OS cells. Conclusion EIF3C may be a promising target for gene therapy of human OsC. However, the precise mechanisms behind the effect of EIF3C on OsC tumorigenesis require further analysis.
Collapse
Affiliation(s)
- Weilu Gao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China,
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China,
| | - Zhengqin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Gongwen Du
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China,
| | - Li Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China,
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China,
| |
Collapse
|
11
|
Shintani T, Higashisaka K, Maeda M, Hamada M, Tsuji R, Kurihara K, Kashiwagi Y, Sato A, Obana M, Yamamoto A, Kawasaki K, Lin Y, Kijima T, Kinehara Y, Miwa Y, Maeda S, Morii E, Kumanogoh A, Tsutsumi Y, Nagatomo I, Fujio Y. Eukaryotic translation initiation factor 3 subunit C is associated with acquired resistance to erlotinib in non-small cell lung cancer. Oncotarget 2018; 9:37520-37533. [PMID: 30680067 PMCID: PMC6331022 DOI: 10.18632/oncotarget.26494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022] Open
Abstract
The acquisition of resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) is one of the major problems in the pharmacotherapy against non-small cell lung cancers; however, molecular mechanisms remain to be fully elucidated. Here, using a newly-established erlotinib-resistant cell line, PC9/ER, from PC9 lung cancer cells, we demonstrated that the expression of translation-related molecules, including eukaryotic translation initiation factor 3 subunit C (eIF3c), was upregulated in PC9/ER cells by proteome analyses. Immunoblot analyses confirmed that eIF3c protein increased in PC9/ER cells, compared with PC9 cells. Importantly, the knockdown of eIF3c with its siRNAs enhanced the drug sensitivity in PC9/ER cells. Mechanistically, we found that LC3B-II was upregulated in PC9/ER cells, while downregulated by the knockdown of eIF3c. Consistently, the overexpression of eIF3c increased the number of autophagosomes, proposing the causality between eIF3c expression and autophagy. Moreover, chloroquine, an autophagy inhibitor, restored the sensitivity to erlotinib. Finally, immunohistochemical analyses of biopsy samples showed that the frequency of eIF3c-positive cases was higher in the patients with EGFR-TKI resistance than those prior to EGFR-TKI treatment. Moreover, the eIF3c-positive cases exhibited poor prognosis in EGFR-TKI treatment. Collectively, the upregulation of eIF3c could impair the sensitivity to EGFR-TKI as a novel mechanism of the drug resistance.
Collapse
Affiliation(s)
- Takuya Shintani
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Kazuma Higashisaka
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Makiko Maeda
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Masaya Hamada
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Ryosuke Tsuji
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Koudai Kurihara
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Yuri Kashiwagi
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Atsuhiro Sato
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Masanori Obana
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Ayaha Yamamoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Keisuke Kawasaki
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ying Lin
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Takashi Kijima
- Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuhei Kinehara
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshihiro Miwa
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Shinichiro Maeda
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,The Center for Advanced Medical Engineering and Informatics, Osaka University, Suita, Japan
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yasushi Fujio
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
12
|
Sriram A, Bohlen J, Teleman AA. Translation acrobatics: how cancer cells exploit alternate modes of translational initiation. EMBO Rep 2018; 19:embr.201845947. [PMID: 30224410 DOI: 10.15252/embr.201845947] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/09/2018] [Accepted: 08/16/2018] [Indexed: 12/11/2022] Open
Abstract
Recent work has brought to light many different mechanisms of translation initiation that function in cells in parallel to canonical cap-dependent initiation. This has important implications for cancer. Canonical cap-dependent translation initiation is inhibited by many stresses such as hypoxia, nutrient limitation, proteotoxic stress, or genotoxic stress. Since cancer cells are often exposed to these stresses, they rely on alternate modes of translation initiation for protein synthesis and cell growth. Cancer mutations are now being identified in components of the translation machinery and in cis-regulatory elements of mRNAs, which both control translation of cancer-relevant genes. In this review, we provide an overview on the various modes of non-canonical translation initiation, such as leaky scanning, translation re-initiation, ribosome shunting, IRES-dependent translation, and m6A-dependent translation, and then discuss the influence of stress on these different modes of translation. Finally, we present examples of how these modes of translation are dysregulated in cancer cells, allowing them to grow, to proliferate, and to survive, thereby highlighting the importance of translational control in cancer.
Collapse
Affiliation(s)
- Ashwin Sriram
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg University, Heidelberg, Germany
| | - Jonathan Bohlen
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg University, Heidelberg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), Heidelberg, Germany .,Heidelberg University, Heidelberg, Germany
| |
Collapse
|
13
|
Wang X, Wang H, Zhao S, Sun P, Wen D, Liu T, Liu H, Yang Z, Ma Z. Eukaryotic translation initiation factor EIF3H potentiates gastric carcinoma cell proliferation. Tissue Cell 2018; 53:23-29. [PMID: 30060823 DOI: 10.1016/j.tice.2018.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/24/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022]
Abstract
Eukaryotic translation initiation factor 3 subunit H (EIF3H) is required for the progression of several types of cancer. However, little is known about the function of EIF3H in gastric carcinoma. To address this issue, in the present study, we investigated EIF3H genetic alterations in and expression of EIF3H in gastric cancer tissue samples using cBioPortal and Oncomine databases. Endogenous EIF3H expression was knocked down in MGC80-3 and AGS gastric cancer cell lines by lentivirus-mediated RNA interference. We confirmed the knockdown efficiency by quantitative real-time PCR and western blotting and evaluated the effects of EIF3H silencing on cell proliferation of gastric cancer with the cell viability and colony formation assays and by flow cytometry. The OncoPrint of EIF3H generated using cBioPortal indicated that EIF3H genetic alterations (mutation, deletion and amplification) were present in two gastric cancer sample sets. The Oncomine analysis revealed that EIF3H mRNA level was upregulated in gastric cancer tissues. EIF3H knockdown inhibited cell proliferation and colony formation in gastric cancer lines and led to cell cycle arrest at the G0/G1 phase, while inducing apoptosis via up- and downregulation of pro- and anti-apoptotic factors, respectively. These results indicate that EIF3H can serve as a novel therapeutic target for the clinical treatment of gastric cancer.
Collapse
Affiliation(s)
- Xudong Wang
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China.
| | - Haizhu Wang
- Geriatric Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Shutao Zhao
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Pengda Sun
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Dacheng Wen
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Tianzhou Liu
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Hao Liu
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Zecheng Yang
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| | - Zhiming Ma
- Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital, Jilin University, Changchun, China
| |
Collapse
|
14
|
Cao K, Arthurs C, Atta-Ul A, Millar M, Beltran M, Neuhaus J, Horn LC, Henrique R, Ahmed A, Thrasivoulou C. Quantitative Analysis of Seven New Prostate Cancer Biomarkers and the Potential Future of the 'Biomarker Laboratory'. Diagnostics (Basel) 2018; 8:diagnostics8030049. [PMID: 30060509 PMCID: PMC6163663 DOI: 10.3390/diagnostics8030049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/11/2018] [Accepted: 07/20/2018] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is the third highest cause of male mortality in the developed world, with the burden of the disease increasing dramatically with demographic change. There are significant limitations to the current diagnostic regimens and no established effective screening modality. To this end, research has discovered hundreds of potential ‘biomarkers’ that may one day be of use in screening, diagnosis or prognostication. However, the barriers to bringing biomarkers to clinical evaluation and eventually into clinical usage have yet to be realised. This is an operational challenge that requires some new thinking and development of paradigms to increase the efficiency of the laboratory process and add ‘value’ to the clinician. Value comes in various forms, whether it be a process that is seamlessly integrated into the hospital laboratory environment or one that can provide additional ‘information’ for the clinical pathologist in terms of risk profiling. We describe, herein, an efficient and tissue-conserving pipeline that uses Tissue Microarrays in a semi-automated process that could, one day, be integrated into the hospital laboratory domain, using seven putative prostate cancer biomarkers for illustration.
Collapse
Affiliation(s)
- Kevin Cao
- Prostate Cancer Research Centre at the Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK.
| | - Callum Arthurs
- Prostate Cancer Research Centre at the Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK.
| | - Ali Atta-Ul
- Prostate Cancer Research Centre, University College London, London WC1E 6BT, UK.
| | - Michael Millar
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH8 9YL, UK.
| | - Mariana Beltran
- Aquila BioMedical, Nine, Edinburgh BioQuarter, 9 Little France Road, Edinburgh EH16 4UX, UK.
| | - Jochen Neuhaus
- Head of Urology Research Laboratories, University of Leipzig, Department of Urology, Research Laboratory, Liebigstr. 19, Building C, 04103 Leipzig, Germany.
| | - Lars-Christian Horn
- Division of Gynecologic, Breast & Perinatal Pathology, University Hospital Leipzig, Liebigstasse 24 D, 04103 Leipzig, Germany.
| | - Rui Henrique
- Department of Pathology, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Abel Salazar Institute of Biomedical Sciences, University of Porto, 4099-002 Porto, Portugal.
| | - Aamir Ahmed
- Prostate Cancer Research Centre at the Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK.
- Prostate Cancer Research Centre, University College London, London WC1E 6BT, UK.
| | - Christopher Thrasivoulou
- Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics, Rockefeller Building, University College London, London WC1E 6BT, UK.
| |
Collapse
|
15
|
Yin Y, Long J, Sun Y, Li H, Jiang E, Zeng C, Zhu W. The function and clinical significance of eIF3 in cancer. Gene 2018; 673:130-133. [PMID: 29908282 DOI: 10.1016/j.gene.2018.06.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/09/2018] [Accepted: 06/11/2018] [Indexed: 11/15/2022]
Abstract
Abnormal regulation of gene expression is essential for tumorigenesis. Several studies indicate that regulation of oncogene expression and neoplastic transformation are controlled by subunits of eukaryotic translation initiation factors (eIFs). Eukaryotic translation initiation factor 3 (eIF3) is the largest (800 kDa) and the most complex mammalian initiation factor. It is composed of 13 non-identical polypeptides designated as eIF3a-m and plays a pivotal role in protein synthesis that bridges the 43S pre-initiation complex and eIF4F-bound mRNA. However, the functional roles of individual subunits are not yet very clear. This review presents on several of aberrant expressed eIF3 subunits which are detected in various human cancers and the associated mechanisms have been acknowledged or are still not sure. Finally, identifying novel targets and biomarkers for caner is of great importance in early diagnosis and treatment of cancer. eIF3 may be a novel target molecule in drug development for cancer treatment and prevention.
Collapse
Affiliation(s)
- Yuting Yin
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Jiali Long
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Yanqin Sun
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Hongmei Li
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Enping Jiang
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Chao Zeng
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| | - Wei Zhu
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| |
Collapse
|
16
|
Cai X, Shen M, Liu X, Nie J. The Possible Role of Eukaryotic Translation Initiation Factor 3 Subunit e (eIF3e) in the Epithelial-Mesenchymal Transition in Adenomyosis. Reprod Sci 2018; 26:377-385. [PMID: 29871559 DOI: 10.1177/1933719118773490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been reported to be involved in adenomyosis by promoting cell invasion and fibrogenesis. But few studies have identified critical factors that regulate EMT process during adenomyosis. The eukaryotic translation initiation factor 3 subunit e (eIF3e) protein is a component of the multisubunit eIF3 complex essential for cap-dependent translation initiation. The aim of this study was to investigate whether eIF3e is involved in EMT in adenomyosis. Ectopic endometrial tissue samples were collected from 40 premenopausal women with ultrasonographically diagnosed and histologically confirmed adenomyosis. As controls, endometrial samples were obtained from 40 cycling premenopausal women patients who underwent surgery for benign gynecologic disorders or cervical intraepithelial neoplasia but without endometriosis, adenomyosis, nor uterine fibroids. All tissue samples were subjected to immunohistochemistry analysis of eIF3e, transforming growth factor-β1 (TGF-β1), E-cadherin, vimentin, Snail, and proliferating cell nuclear antigen (PCNA). The epithelial component of ectopic endometrium showed significantly reduced immunoreactivity against eIF3e and E-cadherin but elevated immunoreactivity against TGF-β1, Snail, vimentin, and PCNA as compared with that of control endometrium (all P values <.05), and the difference was not affected by age, parity, or menstrual phase. The eIF3e staining levels correlated negatively with those of TGF-β1, vimentin, Snail, and PCNA (both P values <.05). These data suggest that decreased eIF3e expression may pave way for EMT in the development of adenomyosis through activating the TGF-β1 signaling pathway. Our study provided novel insights into the development and treatments of adenomyosis.
Collapse
Affiliation(s)
- Xianjun Cai
- 1 Department of Obstetrics and Gynecology, the Seventh People's Hospital, Ningbo, Zhejiang, China
| | - Minhong Shen
- 2 Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- 2 Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jichan Nie
- 2 Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Shirokikh NE, Preiss T. Translation initiation by cap-dependent ribosome recruitment: Recent insights and open questions. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1473. [PMID: 29624880 DOI: 10.1002/wrna.1473] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/02/2018] [Accepted: 02/14/2018] [Indexed: 12/14/2022]
Abstract
Gene expression universally relies on protein synthesis, where ribosomes recognize and decode the messenger RNA template by cycling through translation initiation, elongation, and termination phases. All aspects of translation have been studied for decades using the tools of biochemistry and molecular biology available at the time. Here, we focus on the mechanism of translation initiation in eukaryotes, which is remarkably more complex than prokaryotic initiation and is the target of multiple types of regulatory intervention. The "consensus" model, featuring cap-dependent ribosome entry and scanning of mRNA leader sequences, represents the predominantly utilized initiation pathway across eukaryotes, although several variations of the model and alternative initiation mechanisms are also known. Recent advances in structural biology techniques have enabled remarkable molecular-level insights into the functional states of eukaryotic ribosomes, including a range of ribosomal complexes with different combinations of translation initiation factors that are thought to represent bona fide intermediates of the initiation process. Similarly, high-throughput sequencing-based ribosome profiling or "footprinting" approaches have allowed much progress in understanding the elongation phase of translation, and variants of them are beginning to reveal the remaining mysteries of initiation, as well as aspects of translation termination and ribosomal recycling. A current view on the eukaryotic initiation mechanism is presented here with an emphasis on how recent structural and footprinting results underpin axioms of the consensus model. Along the way, we further outline some contested mechanistic issues and major open questions still to be addressed. This article is categorized under: Translation > Translation Mechanisms Translation > Translation Regulation RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Nikolay E Shirokikh
- EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Thomas Preiss
- EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| |
Collapse
|
18
|
Lacerda R, Menezes J, Romão L. More than just scanning: the importance of cap-independent mRNA translation initiation for cellular stress response and cancer. Cell Mol Life Sci 2017; 74:1659-1680. [PMID: 27913822 PMCID: PMC11107732 DOI: 10.1007/s00018-016-2428-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/24/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022]
Abstract
The scanning model for eukaryotic mRNA translation initiation states that the small ribosomal subunit, along with initiation factors, binds at the cap structure at the 5' end of the mRNA and scans the 5' untranslated region (5'UTR) until an initiation codon is found. However, under conditions that impair canonical cap-dependent translation, the synthesis of some proteins is kept by alternative mechanisms that are required for cell survival and stress recovery. Alternative modes of translation initiation include cap- and/or scanning-independent mechanisms of ribosomal recruitment. In most cap-independent translation initiation events there is a direct recruitment of the 40S ribosome into a position upstream, or directly at, the initiation codon via a specific internal ribosome entry site (IRES) element in the 5'UTR. Yet, in some cellular mRNAs, a different translation initiation mechanism that is neither cap- nor IRES-dependent seems to occur through a special RNA structure called cap-independent translational enhancer (CITE). Recent evidence uncovered a distinct mechanism through which mRNAs containing N 6-methyladenosine (m6A) residues in their 5'UTR directly bind eukaryotic initiation factor 3 (eIF3) and the 40S ribosomal subunit in order to initiate translation in the absence of the cap-binding proteins. This review focuses on the important role of cap-independent translation mechanisms in human cells and how these alternative mechanisms can either act individually or cooperate with other cis-acting RNA regulons to orchestrate specific translational responses triggered upon several cellular stress states, and diseases such as cancer. Elucidation of these non-canonical mechanisms reveals the complexity of translational control and points out their potential as prospective novel therapeutic targets.
Collapse
Affiliation(s)
- Rafaela Lacerda
- Department of Human Genetics, Instituto Nacional de Saúde Doutor Ricardo Jorge, Av. Padre Cruz, 1649-016, Lisbon, Portugal
- Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Juliane Menezes
- Department of Human Genetics, Instituto Nacional de Saúde Doutor Ricardo Jorge, Av. Padre Cruz, 1649-016, Lisbon, Portugal
- Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Luísa Romão
- Department of Human Genetics, Instituto Nacional de Saúde Doutor Ricardo Jorge, Av. Padre Cruz, 1649-016, Lisbon, Portugal.
- Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
19
|
eIF3f reduces tumor growth by directly interrupting clusterin with anti-apoptotic property in cancer cells. Oncotarget 2017; 7:18541-57. [PMID: 26988917 PMCID: PMC4951308 DOI: 10.18632/oncotarget.8105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 02/05/2016] [Indexed: 01/07/2023] Open
Abstract
Clusterin is a secretory heterodimeric glycoprotein and the overexpression of secretory clusterin (sCLU) promotes cancer cell proliferation and reduces chemosensitivity. Therefore, sCLU might be an effective target for anticancer therapy. In the current study, we identified eIF3f as a novel CLU-interacting protein and demonstrated its novel function as a CLU inhibitor. The overexpression of eIF3f retarded cancer cell growth significantly and induced apoptosis. In addition, eIF3f interacted with the α-chain (1–227) of sCLU. This interaction blocked modification of psCLU, thereby decreasing the expression and secretion of α/β CLU. Consequently, the overexpression of eIF3f suppressed Akt and ERK signaling and subsequently depleted CLU expression. In addition, eIF3F stabilized p53, which increased the expression of p21 and Bax. Interestingly, the expression of Bax was increased without the activation of p53. eIF3f injected into a xenograft model of human cervical cancer in nude mice markedly inhibited tumor growth. The identification of this novel function of eIF3f as a sCLU inhibitor might open novel avenues for developing improved strategies for CLU-targeted anti-cancer therapies.
Collapse
|
20
|
Assembly of eIF3 Mediated by Mutually Dependent Subunit Insertion. Structure 2016; 24:886-96. [PMID: 27210288 DOI: 10.1016/j.str.2016.02.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/19/2016] [Accepted: 02/21/2016] [Indexed: 02/05/2023]
Abstract
Eukaryotic initiation factor 3 (eIF3), an essential multi-protein complex involved in translation initiation, is composed of 12 tightly associated subunits in humans. While the overall structure of eIF3 is known, the mechanism of its assembly and structural consequences of dysregulation of eIF3 subunit expression seen in many cancers is largely unknown. Here we show that subunits in eIF3 assemble into eIF3 in an interdependent manner. Assembly of eIF3 is governed primarily by formation of a helical bundle, composed of helices extending C-terminally from PCI-MPN domains in eight subunits. We propose that, while the minimal subcomplex of human-like eIF3 functional for translation initiation in cells consists of subunits a, b, c, f, g, i, and m, numerous other eIF3 subcomplexes exist under circumstances of subunit over- or underexpression. Thus, eIF3 subcomplexes formed or "released" due to dysregulated subunit expression may be determining factors contributing to eIF3-related cancers.
Collapse
|
21
|
Zheng Q, Liu H, Ye J, Zhang H, Jia Z, Cao J. Nuclear distribution of eIF3g and its interacting nuclear proteins in breast cancer cells. Mol Med Rep 2016; 13:2973-80. [PMID: 26935993 PMCID: PMC4805062 DOI: 10.3892/mmr.2016.4935] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 01/11/2016] [Indexed: 12/11/2022] Open
Abstract
Eukaryotic translation initiation factor 3 subunit g (eIF3g) is a core subunit of the eukaryotic translation initiation factor 3 complex, and is important in the initiation of translation. It is also involved in caspase-mediated apoptosis, and is upregulated in multidrug-resistant cancer cells. In the present study, the nuclear distribution of eIF3g was determined by performing co-immunoprecipitation of proteins that potentially interact with eIF3g in the nucleus. Mass spectrometry characterization showed that three proteins, heterogeneous nuclear ribonucleoprotein U/scaffold attachment factor A, HSZFP36/zinc finger protein 823 and β-actin, were among the candidate eIF3g-interacting proteins in the nucleus. The protein-protein interaction was further confirmed by cross-linking and a glutathione S-transferase pull-down assay, followed by western blotting. The co-localization of these proteins was determined by confocal microscopy. These findings provide novel insight into the possible functions of eIF3g in the nucleus and serves as an important first step for further investigation of the roles of eIF3g in cancer development.
Collapse
Affiliation(s)
- Qiaoli Zheng
- Clinical Research Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Hao Liu
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310007, P.R. China
| | - Jingjia Ye
- Clinical Research Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Hui Zhang
- Department of Clinical Laboratory, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhenyu Jia
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310007, P.R. China
| | - Jiang Cao
- Clinical Research Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
22
|
Yu J, Lan X, Chen X, Yu C, Xu Y, Liu Y, Xu L, Fan HY, Tong C. Protein synthesis and degradation are critical to regulate germline stem cell homeostasis in Drosophila testes. Development 2016; 143:2930-45. [DOI: 10.1242/dev.134247] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/11/2016] [Indexed: 12/14/2022]
Abstract
The homeostasis of self-renewal and differentiation in stem cells is strictly controlled by intrinsic signals and their niche. We conducted a large-scale RNA interference (RNAi) screen in Drosophila testes and identified 221 genes required for germline stem cell (GSC) maintenance or differentiation. Knockdown of these genes in transit-amplifying spermatogonia and cyst cells further revealed various phenotypes. Complex analysis uncovered that many of the identified genes are involved in key steps of protein synthesis and degradation. A group of genes that are required for mRNA splicing and protein translation contributes to both GSC self-renewal and early germ cell differentiation. Loss of genes in protein degradation pathway in cyst cells leads to testis tumor with overproliferated germ cells. Importantly, in the Cullin 4-Ring E3 ubiquitin ligase (CRL4) complex, we identified multiple proteins that are critical to GSC self-renewal. pic/DDB1, the linker protein of CRL4, is not only required for GSC self-renewal in flies but also for maintenance of spermatogonial stem cells (SSCs) in mice.
Collapse
Affiliation(s)
- Jun Yu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
- Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Xiang Lan
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Xia Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Chao Yu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Yiwen Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Yujuan Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
- Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Lingna Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Heng-Yu Fan
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Chao Tong
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
23
|
Biological insights into the expression of translation initiation factors from recombinant CHOK1SV cell lines and their relationship to enhanced productivity. Biochem J 2015; 472:261-73. [DOI: 10.1042/bj20150928] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/29/2015] [Indexed: 12/31/2022]
Abstract
We show for translation initiation factors involved in formation of the closed loop mRNA, their expression is associated with recombinant antibody productivity in Chinese hamster ovary cells and maintaining these is important in determining the cells capacity for antibody productivity.
Collapse
|
24
|
Chen M, Zhu A, Storey KB. Comparative phosphoproteomic analysis of intestinal phosphorylated proteins in active versus aestivating sea cucumbers. J Proteomics 2015; 135:141-150. [PMID: 26385000 DOI: 10.1016/j.jprot.2015.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/27/2015] [Accepted: 09/09/2015] [Indexed: 01/18/2023]
Abstract
UNLABELLED The sea cucumber Apostichopus japonicus is becoming an excellent model marine invertebrate for studies of environmentally-induced aestivation. Reversible protein phosphorylation as a regulatory mechanism in aestivation is known for some terrestrial aestivators but has never before been documented in sea cucumbers. The present study provides a global quantitative analysis of the role of reversible phosphorylation in sea cucumber aestivation by using tandem mass tag (TMT) labeling followed by an IMAC enrichment strategy to map aestivation-responsive changes in the phosphoproteome of sea cucumber intestine. We identified 2295 unique phosphosites derived from 1283 phosphoproteins and, of these, 211 hyperphosphorylated and 65 hypophosphorylated phosphoproteins were identified in intestine during deep aestivation compared with the active state based on the following criterion: quantitative ratios over 1.5 or less than 0.67 with corrected p-value <0.05. Six major functional classes of proteins exhibited changes in their phosphorylation status during aestivation: (1) protein synthesis, (2) transcriptional regulators, (3) kinases, (4) signaling, (5) transporter, (6) DNA binding. These data on the global involvement of phosphorylation in sea cucumber aestivation significantly improve our understanding of the regulatory mechanisms involved in metabolic arrest when marine invertebrates face environmental stress and provide substantial candidate phosphorylated proteins that could be important for identifying functionally adaptive variation in marine invertebrates. SIGNIFICANCE Sea cucumber Apostichopus japonicus is an excellent model organism for studies of environmentally-induced aestivation by a marine invertebrate. The present study provides the first quantitative phosphoproteomic analysis of sea cucumber aestivation using isobaric tag based TMT labeling followed by an IMAC enrichment strategy. These data on the global involvement of phosphorylation in sea cucumber aestivation significantly improve our understanding of the regulatory mechanism involved in metabolic arrest when marine invertebrates face environmental stress and provide substantial candidate phosphorylated proteins that could be important for identifying functionally adaptive variation in marine invertebrates. This study also demonstrates the usefulness of the TMT-based quantitative phosphoproteomics approach to explore the survival responses of a non-model marine invertebrate species to seasonal changes in its environment.
Collapse
Affiliation(s)
- Muyan Chen
- Fisheries College, Ocean University of China, Qingdao, PR China.
| | - Aijun Zhu
- Fisheries College, Ocean University of China, Qingdao, PR China
| | - Kenneth B Storey
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON, Canada, K1S 5B6
| |
Collapse
|
25
|
Borgo C, Franchin C, Salizzato V, Cesaro L, Arrigoni G, Matricardi L, Pinna LA, Donella-Deana A. Protein kinase CK2 potentiates translation efficiency by phosphorylating eIF3j at Ser127. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1693-701. [PMID: 25887626 DOI: 10.1016/j.bbamcr.2015.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/17/2015] [Accepted: 04/07/2015] [Indexed: 11/18/2022]
Abstract
In eukaryotic protein synthesis the translation initiation factor 3 (eIF3) is a key player in the recruitment and assembly of the translation initiation machinery. Mammalian eIF3 consists of 13 subunits, including the loosely associated eIF3j subunit that plays a stabilizing role in the eIF3 complex formation and interaction with the 40S ribosomal subunit. By means of both co-immunoprecipitation and mass spectrometry analyses we demonstrate that the protein kinase CK2 interacts with and phosphorylates eIF3j at Ser127. Inhibition of CK2 activity by CX-4945 or down-regulation of the expression of CK2 catalytic subunit by siRNA cause the dissociation of j-subunit from the eIF3 complex as judged from glycerol gradient sedimentation. This finding proves that CK2-phosphorylation of eIF3j is a prerequisite for its association with the eIF3 complex. Expression of Ser127Ala-eIF3j mutant impairs both the interaction of mutated j-subunit with the other eIF3 subunits and the overall protein synthesis. Taken together our data demonstrate that CK2-phosphorylation of eIF3j at Ser127 promotes the assembly of the eIF3 complex, a crucial step in the activation of the translation initiation machinery.
Collapse
Affiliation(s)
- Christian Borgo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy; CNR Institute of NeuroSciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy
| | - Cinzia Franchin
- Proteomic Center of Padova University, Via G. Orus B2, 35129 Padova, Italy
| | - Valentina Salizzato
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy; CNR Institute of NeuroSciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy
| | - Luca Cesaro
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy; CNR Institute of NeuroSciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy
| | - Giorgio Arrigoni
- Proteomic Center of Padova University, Via G. Orus B2, 35129 Padova, Italy
| | - Laura Matricardi
- Venitian Institute of Oncology (IOV-IRCCS), Via Gattamelata 64, 35128 Padova, Italy
| | - Lorenzo A Pinna
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy; CNR Institute of NeuroSciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy
| | - Arianna Donella-Deana
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy; CNR Institute of NeuroSciences, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy.
| |
Collapse
|
26
|
eIF3 targets cell-proliferation messenger RNAs for translational activation or repression. Nature 2015; 522:111-4. [PMID: 25849773 PMCID: PMC4603833 DOI: 10.1038/nature14267] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/27/2015] [Indexed: 01/23/2023]
Abstract
Regulation of protein synthesis is fundamental for all aspects of eukaryotic biology by controlling development, homeostasis and stress responses. The 13-subunit, 800-kilodalton eukaryotic initiation factor 3 (eIF3) organizes initiation factor and ribosome interactions required for productive translation. However, current understanding of eIF3 function does not explain genetic evidence correlating eIF3 deregulation with tissue-specific cancers and developmental defects. Here we report the genome-wide discovery of human transcripts that interact with eIF3 using photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP). eIF3 binds to a highly specific program of messenger RNAs involved in cell growth control processes, including cell cycling, differentiation and apoptosis, via the mRNA 5' untranslated region. Surprisingly, functional analysis of the interaction between eIF3 and two mRNAs encoding the cell proliferation regulators c-JUN and BTG1 reveals that eIF3 uses different modes of RNA stem-loop binding to exert either translational activation or repression. Our findings illuminate a new role for eIF3 in governing a specialized repertoire of gene expression and suggest that binding of eIF3 to specific mRNAs could be targeted to control carcinogenesis.
Collapse
|
27
|
HAO JINMIN, WANG ZHIMING, WANG YAOWU, LIANG ZHAOHUI, ZHANG XIN, ZHAO ZONGMAO, JIAO BAOHUA. Eukaryotic initiation factor 3C silencing inhibits cell proliferation and promotes apoptosis in human glioma. Oncol Rep 2015; 33:2954-62. [DOI: 10.3892/or.2015.3881] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/09/2015] [Indexed: 11/05/2022] Open
|
28
|
Galligan J, Martinez-Noël G, Arndt V, Hayes S, Chittenden TW, Harper JW, Howley PM. Proteomic analysis and identification of cellular interactors of the giant ubiquitin ligase HERC2. J Proteome Res 2015; 14:953-66. [PMID: 25476789 PMCID: PMC4324439 DOI: 10.1021/pr501005v] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Indexed: 01/10/2023]
Abstract
HERC2 is a large E3 ubiquitin ligase with multiple structural domains that has been implicated in an array of cellular processes. Mutations in HERC2 are linked to developmental delays and impairment caused by nervous system dysfunction, such as Angelman Syndrome and autism-spectrum disorders. However, HERC2 cellular activity and regulation remain poorly understood. We used a broad proteomic approach to survey the landscape of cellular proteins that interact with HERC2. We identified nearly 300 potential interactors, a subset of which we validated binding to HERC2. The potential HERC2 interactors included the eukaryotic translation initiation factor 3 complex, the intracellular transport COPI coatomer complex, the glycogen regulator phosphorylase kinase, beta-catenin, PI3 kinase, and proteins involved in fatty acid transport and iron homeostasis. Through a complex bioinformatic analysis of potential interactors, we linked HERC2 to cellular processes including intracellular protein trafficking and transport, metabolism of cellular energy, and protein translation. Given its size, multidomain structure, and association with various cellular activities, HERC2 may function as a scaffold to integrate protein complexes and bridge critical cellular pathways. This work provides a significant resource with which to interrogate HERC2 function more deeply and evaluate its contributions to mechanisms governing cellular homeostasis and disease.
Collapse
Affiliation(s)
- Jeffrey
T. Galligan
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Gustavo Martinez-Noël
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Verena Arndt
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Sebastian Hayes
- Department
of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Thomas W. Chittenden
- Research
Computing Group, Harvard Medical School, 25 Shattuck Street #500, Boston, Massachusetts 02115, United States
- Complex Biological
Systems Alliance, 17 Peterson Road, North Andover, Massachusetts 01845, United States
| | - J. Wade Harper
- Department
of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Peter M. Howley
- Department
of Microbiology and Immunobiology, Harvard
Medical School, 77 Avenue
Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
29
|
Gao B, Roux PP. Translational control by oncogenic signaling pathways. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:753-65. [PMID: 25477072 DOI: 10.1016/j.bbagrm.2014.11.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 01/04/2023]
Abstract
Messenger RNA (mRNA) translation is highly regulated in cells and plays an integral role in the overall process of gene expression. The initiation phase of translation is considered to be the most rate-limiting and is often targeted by oncogenic signaling pathways to promote global protein synthesis and the selective translation of tumor-promoting mRNAs. Translational control is a crucial component of cancer development as it allows cancer cells to adapt to the altered metabolism that is generally associated with the tumor state. The phosphoinositide 3-kinase (PI3K)/Akt and Ras/mitogen-activated protein kinase (MAPK) pathways are strongly implicated in cancer etiology, and they exert their biological effects by modulating both global and specific mRNA translation. In addition to having respective translational targets, these pathways also impinge on the mechanistic/mammalian target of rapamycin (mTOR), which acts as a critical signaling node linking nutrient sensing to the coordinated regulation of cellular metabolism. mTOR is best known as a central regulator of protein synthesis and has been implicated in an increasing number of pathological conditions, including cancer. In this article, we describe the current knowledge on the roles and regulation of mRNA translation by various oncogenic signaling pathways, as well as the relevance of these molecular mechanisms to human malignancies. This article is part of a Special Issue entitled: Translation and cancer.
Collapse
Affiliation(s)
- Beichen Gao
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Québec, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
30
|
Yue MM, Lv K, Meredith SC, Martindale JL, Gorospe M, Schuger L. Novel RNA-binding protein P311 binds eukaryotic translation initiation factor 3 subunit b (eIF3b) to promote translation of transforming growth factor β1-3 (TGF-β1-3). J Biol Chem 2014; 289:33971-83. [PMID: 25336651 DOI: 10.1074/jbc.m114.609495] [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: 11/06/2022] Open
Abstract
P311, a conserved 8-kDa intracellular protein expressed in brain, smooth muscle, regenerating tissues, and malignant glioblastomas, represents the first documented stimulator of TGF-β1-3 translation in vitro and in vivo. Here we initiated efforts to define the mechanism underlying P311 function. PONDR® (Predictor Of Naturally Disordered Regions) analysis suggested and CD confirmed that P311 is an intrinsically disordered protein, therefore requiring an interacting partner to acquire tertiary structure and function. Immunoprecipitation coupled with mass spectroscopy identified eIF3 subunit b (eIF3b) as a novel P311 binding partner. Immunohistochemical colocalization, GST pulldown, and surface plasmon resonance studies revealed that P311-eIF3b interaction is direct and has a Kd of 1.26 μm. Binding sites were mapped to the non-canonical RNA recognition motif of eIF3b and a central 11-amino acid-long region of P311, here referred to as eIF3b binding motif. Disruption of P311-eIF3b binding inhibited translation of TGF-β1, 2, and 3, as indicated by luciferase reporter assays, polysome fractionation studies, and Western blot analysis. RNA precipitation assays after UV cross-linking and RNA-protein EMSA demonstrated that P311 binds directly to TGF-β 5'UTRs mRNAs through a previously unidentified RNA recognition motif-like motif. Our results demonstrate that P311 is a novel RNA-binding protein that, by interacting with TGF-βs 5'UTRs and eIF3b, stimulates the translation of TGF-β1, 2, and 3.
Collapse
Affiliation(s)
| | | | - Stephen C Meredith
- From the Departments of Pathology and Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637 and
| | - Jennifer L Martindale
- the Laboratory of Genetics, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Myriam Gorospe
- the Laboratory of Genetics, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | | |
Collapse
|
31
|
Int6/eIF3e is essential for proliferation and survival of human glioblastoma cells. Int J Mol Sci 2014; 15:2172-90. [PMID: 24481065 PMCID: PMC3958844 DOI: 10.3390/ijms15022172] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 12/25/2013] [Accepted: 01/23/2014] [Indexed: 12/15/2022] Open
Abstract
Glioblastomas (GBM) are very aggressive and malignant brain tumors, with frequent relapses despite an appropriate treatment combining surgery, chemotherapy and radiotherapy. In GBM, hypoxia is a characteristic feature and activation of Hypoxia Inducible Factors (HIF-1α and HIF-2α) has been associated with resistance to anti-cancer therapeutics. Int6, also named eIF3e, is the “e” subunit of the translation initiation factor eIF3, and was identified as novel regulator of HIF-2α. Eukaryotic initiation factors (eIFs) are key factors regulating total protein synthesis, which controls cell growth, size and proliferation. The functional significance of Int6 and the effect of Int6/EIF3E gene silencing on human brain GBM has not yet been described and its role on the HIFs is unknown in glioma cells. In the present study, we show that Int6/eIF3e suppression affects cell proliferation, cell cycle and apoptosis of various GBM cells. We highlight that Int6 inhibition induces a diminution of proliferation through cell cycle arrest and increased apoptosis. Surprisingly, these phenotypes are independent of global cell translation inhibition and are accompanied by decreased HIF expression when Int6 is silenced. In conclusion, we demonstrate here that Int6/eIF3e is essential for proliferation and survival of GBM cells, presumably through modulation of the HIFs.
Collapse
|
32
|
Smith MD, Gu Y, Querol-Audí J, Vogan JM, Nitido A, Cate JHD. Human-like eukaryotic translation initiation factor 3 from Neurospora crassa. PLoS One 2013; 8:e78715. [PMID: 24250809 PMCID: PMC3826745 DOI: 10.1371/journal.pone.0078715] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 09/22/2013] [Indexed: 02/05/2023] Open
Abstract
Eukaryotic translation initiation factor 3 (eIF3) is a key regulator of translation initiation, but its in vivo assembly and molecular functions remain unclear. Here we show that eIF3 from Neurospora crassa is structurally and compositionally similar to human eIF3. N. crassa eIF3 forms a stable 12-subunit complex linked genetically and biochemically to the 13th subunit, eIF3j, which in humans modulates mRNA start codon selection. Based on N. crassa genetic analysis, most subunits in eIF3 are essential. Subunits that can be deleted (e, h, k and l) map to the right side of the eIF3 complex, suggesting that they may coordinately regulate eIF3 function. Consistent with this model, subunits eIF3k and eIF3l are incorporated into the eIF3 complex as a pair, and their insertion depends on the presence of subunit eIF3h, a key regulator of vertebrate development. Comparisons to other eIF3 complexes suggest that eIF3 assembles around an eIF3a and eIF3c dimer, which may explain the coordinated regulation of human eIF3 levels. Taken together, these results show that Neurospora crassa eIF3 provides a tractable system for probing the structure and function of human-like eIF3 in the context of living cells.
Collapse
Affiliation(s)
- M. Duane Smith
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Yu Gu
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Jordi Querol-Audí
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Jacob M. Vogan
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Adam Nitido
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Jamie H. D. Cate
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- Department of Chemistry, University of California, Berkeley, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Xia Z, Tong X, Liang F, Zhang Y, Kuok C, Zhang Y, Liu X, Zhu Z, Lin S, Zhang B. Eif3ba regulates cranial neural crest development by modulating p53 in zebrafish. Dev Biol 2013; 381:83-96. [PMID: 23791820 DOI: 10.1016/j.ydbio.2013.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/01/2013] [Accepted: 06/06/2013] [Indexed: 02/05/2023]
Abstract
Congenital diseases caused by abnormal development of the cranial neural crest usually present craniofacial malformations and heart defects while the precise mechanism is not fully understood. Here, we show that the zebrafish eif3ba mutant caused by pseudo-typed retrovirus insertion exhibited a similar phenotype due to the hypogenesis of cranial neural crest cells (NCCs). The derivatives of cranial NCCs, including the NCC-derived cell population of pharyngeal arches, craniofacial cartilage, pigment cells and the myocardium derived from cardiac NCCs, were affected in this mutant. The expression of several neural crest marker genes, including crestin, dlx2a and nrp2b, was specifically reduced in the cranial regions of the eif3ba mutant. Through fluorescence-tracing of the cranial NCC migration marker nrp2b, we observed reduced intensity of NCC-derived cells in the heart. In addition, p53 was markedly up-regulated in the eif3ba mutant embryos, which correlated with pronounced apoptosis in the cranial area as shown by TUNEL staining. These findings suggest a novel function of eif3ba during embryonic development and a novel level of regulation in the process of cranial NCC development, in addition to providing a potential animal model to mimic congenital diseases due to cranial NCC defects. Furthermore, we report the identification of a novel transgenic fish line Et(gata2a:EGFP)pku418 to trace the migration of cranial NCCs (including cardiac NCCs); this may serve as an invaluable tool for investigating the development and dynamics of cranial NCCs during zebrafish embryogenesis.
Collapse
Affiliation(s)
- Zhidan Xia
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The translational factor eIF3f: the ambivalent eIF3 subunit. Cell Mol Life Sci 2013; 70:3603-16. [PMID: 23354061 PMCID: PMC3771369 DOI: 10.1007/s00018-013-1263-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/18/2012] [Accepted: 01/07/2013] [Indexed: 11/15/2022]
Abstract
The regulation of the protein synthesis has a crucial role in governing the eukaryotic cell growth. Subtle changes of proteins involved in the translation process may alter the rate of the protein synthesis and modify the cell fate by shifting the balance from normal status into a tumoral or apoptotic one. The largest eukaryotic initiation factor involved in translation regulation is eIF3. Amongst the 13 factors constituting eIF3, the f subunit finely regulates this balance in a cell-type-specific manner. Loss of this factor causes malignancy in several cells, and atrophy in normal muscle cells. The intracellular interacting partners which influence its physiological significance in both cancer and muscle cells are detailed in this review. By delineating the global interaction network of this factor and by clarifying its intracellular role, it becomes apparent that the f subunit represents a promising candidate molecule to use for biotherapeutic applications.
Collapse
|
35
|
Neusiedler J, Mocquet V, Limousin T, Ohlmann T, Morris C, Jalinot P. INT6 interacts with MIF4GD/SLIP1 and is necessary for efficient histone mRNA translation. RNA (NEW YORK, N.Y.) 2012; 18:1163-1177. [PMID: 22532700 PMCID: PMC3358639 DOI: 10.1261/rna.032631.112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/06/2012] [Indexed: 05/31/2023]
Abstract
The INT6/EIF3E protein has been implicated in mouse and human breast carcinogenesis. This subunit of the eIF3 translation initiation factor that includes a PCI domain exhibits specific features such as presence in the nucleus and ability to interact with other important cellular protein complexes like the 26S proteasome and the COP9 signalosome. It has been previously shown that INT6 was not essential for bulk translation, and this protein is considered to regulate expression of specific mRNAs. Based on the results of a two-hybrid screen performed with INT6 as bait, we characterize in this article the MIF4GD/SLIP1 protein as an interactor of this eIF3 subunit. MIF4GD was previously shown to associate with SLBP, which binds the stem-loop located at the 3' end of the histone mRNAs, and to be necessary for efficient translation of these cell cycle-regulated mRNAs that lack a poly(A) tail. In line with the interaction of both proteins, we show using the RNA interference approach that INT6 is also essential to S-phase histone mRNA translation. This was observed by analyzing expression of endogenous histones and by testing heterologous constructs placing the luciferase reporter gene under the control of the stem-loop element of various histone genes. With such a reporter plasmid, silencing and overexpression of INT6 exerted opposite effects. In agreement with these results, INT6 and MIF4GD were observed to colocalize in cytoplasmic foci. We conclude from these data that INT6, by establishing interactions with MIF4GD and SLBP, plays an important role in translation of poly(A) minus histone mRNAs.
Collapse
Affiliation(s)
- Julia Neusiedler
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| | - Vincent Mocquet
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| | - Taran Limousin
- Virologie Humaine, Unité 758, Institut National de la Santé et de la Recherche Médicale, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| | - Theophile Ohlmann
- Virologie Humaine, Unité 758, Institut National de la Santé et de la Recherche Médicale, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| | - Christelle Morris
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| | - Pierre Jalinot
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| |
Collapse
|
36
|
Overexpression of eIF3a in Squamous Cell Carcinoma of the Oral Cavity and Its Putative Relation to Chemotherapy Response. JOURNAL OF ONCOLOGY 2012; 2012:901956. [PMID: 22619676 PMCID: PMC3347757 DOI: 10.1155/2012/901956] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 02/10/2012] [Indexed: 01/23/2023]
Abstract
The eukaryotic translation initiation factor eIF3a is one of the core subunits of the translation initiation complex eIF3, responsible for ribosomal subunit joining and mRNA recruitment to the ribosome. It is known to play an important role in general translation initiation as well as in the specific translational regulation of various gene products, among which many influence tumour development, progression, and the therapeutically important pathways of DNA damage repair. Therefore, beyond its role in protein synthesis, eIF3a is emerging as regulator in tumour pathogenesis and therapy response and, therefore, a potential tumor marker. By means of a tissue microarray (TMA) for histopathological and statistical assessment, we here show eIF3a expression in 103 cases of squamous cell carcinoma of the oral cavity (OSCC), representing tissues from 103 independent patients. A subset of the study cohort was treated with platinum based therapy. Our results show that the 170 kDa protein is upregulated in OSCC and correlates with good overall survival. Overexpressing tumors respond better to platinum-based chemotherapy, suggesting eIF3a as a putative predictive as well as prognostic tumor marker in OSCC.
Collapse
|
37
|
Choe J, Oh N, Park S, Lee YK, Song OK, Locker N, Chi SG, Kim YK. Translation initiation on mRNAs bound by nuclear cap-binding protein complex CBP80/20 requires interaction between CBP80/20-dependent translation initiation factor and eukaryotic translation initiation factor 3g. J Biol Chem 2012; 287:18500-9. [PMID: 22493286 DOI: 10.1074/jbc.m111.327528] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the cytoplasm of mammalian cells, either cap-binding proteins 80 and 20 (CBP80/20) or eukaryotic translation initiation factor (eIF) 4E can direct the initiation of translation. Although the recruitment of ribosomes to mRNAs during eIF4E-dependent translation (ET) is well characterized, the molecular mechanism for CBP80/20-dependent translation (CT) remains obscure. Here, we show that CBP80/20-dependent translation initiation factor (CTIF), which has been shown to be preferentially involved in CT but not ET, specifically interacts with eIF3g, a component of the eIF3 complex involved in ribosome recruitment. By interacting with eIF3g, CTIF serves as an adaptor protein to bridge the CBP80/20 and the eIF3 complex, leading to efficient ribosome recruitment during CT. Accordingly, down-regulation of CTIF using a small interfering RNA causes a redistribution of CBP80 from polysome fractions to subpolysome fractions, without significant consequence to eIF4E distribution. In addition, down-regulation of eIF3g inhibits the efficiency of nonsense-mediated mRNA decay, which is tightly coupled to CT but not to ET. Moreover, the artificial tethering of CTIF to an intercistronic region of dicistronic mRNA results in translation of the downstream cistron in an eIF3-dependent manner. These findings support the idea that CT mechanistically differs from ET.
Collapse
Affiliation(s)
- Junho Choe
- School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Koc EC, Koc H. Regulation of mammalian mitochondrial translation by post-translational modifications. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:1055-66. [PMID: 22480953 DOI: 10.1016/j.bbagrm.2012.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 01/24/2012] [Accepted: 03/16/2012] [Indexed: 11/29/2022]
Abstract
Mitochondria are responsible for the production of over 90% of the energy in eukaryotes through oxidative phosphorylation performed by electron transfer and ATP synthase complexes. Mitochondrial translation machinery is responsible for the synthesis of 13 essential proteins of these complexes encoded by the mitochondrial genome. Emerging data suggest that acetyl-CoA, NAD(+), and ATP are involved in regulation of this machinery through post-translational modifications of its protein components. Recent high-throughput proteomics analyses and mapping studies have provided further evidence for phosphorylation and acetylation of ribosomal proteins and translation factors. Here, we will review our current knowledge related to these modifications and their possible role(s) in the regulation of mitochondrial protein synthesis using the homology between mitochondrial and bacterial translation machineries. However, we have yet to determine the effects of phosphorylation and acetylation of translation components in mammalian mitochondrial biogenesis. This article is part of a Special Issue entitled: Mitochondrial Gene Expression.
Collapse
Affiliation(s)
- Emine C Koc
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV 25755, USA.
| | | |
Collapse
|
39
|
Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J 2012; 441:1-21. [PMID: 22168436 DOI: 10.1042/bj20110892] [Citation(s) in RCA: 767] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The ribosomal protein S6K (S6 kinase) represents an extensively studied effector of the TORC1 [TOR (target of rapamycin) complex 1], which possesses important yet incompletely defined roles in cellular and organismal physiology. TORC1 functions as an environmental sensor by integrating signals derived from diverse environmental cues to promote anabolic and inhibit catabolic cellular functions. mTORC1 (mammalian TORC1) phosphorylates and activates S6K1 and S6K2, whose first identified substrate was rpS6 (ribosomal protein S6), a component of the 40S ribosome. Studies over the past decade have uncovered a number of additional S6K1 substrates, revealing multiple levels at which the mTORC1-S6K1 axis regulates cell physiology. The results thus far indicate that the mTORC1-S6K1 axis controls fundamental cellular processes, including transcription, translation, protein and lipid synthesis, cell growth/size and cell metabolism. In the present review we summarize the regulation of S6Ks, their cellular substrates and functions, and their integration within rapidly expanding mTOR (mammalian TOR) signalling networks. Although our understanding of the role of mTORC1-S6K1 signalling in physiology remains in its infancy, evidence indicates that this signalling axis controls, at least in part, glucose homoeostasis, insulin sensitivity, adipocyte metabolism, body mass and energy balance, tissue and organ size, learning, memory and aging. As dysregulation of this signalling axis contributes to diverse disease states, improved understanding of S6K regulation and function within mTOR signalling networks may enable the development of novel therapeutics.
Collapse
|