1
|
Roshan P, Biswas A, Ahmed S, Anagnos S, Luebbers R, Harish K, Li M, Nguyen N, Zhou G, Tedeschi F, Hathuc V, Lin Z, Hamilton Z, Origanti S. Sequestration of ribosomal subunits as inactive 80S by targeting eIF6 limits mitotic exit and cancer progression. Nucleic Acids Res 2025; 53:gkae1272. [PMID: 39727167 PMCID: PMC11879136 DOI: 10.1093/nar/gkae1272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/25/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024] Open
Abstract
Moderating the pool of active ribosomal subunits is critical for maintaining global translation rates. A factor crucial for modulating the 60S ribosomal subunit is eukaryotic translation initiation factor-6 (eIF6). Release of eIF6 from the 60S subunit is essential to permit 60S interactions with the 40S subunit. Here, using the eIF6-N106S mutant, we show that disrupting eIF6 interaction with the 60S subunit leads to an increase in vacant 80S ribosomes. It further highlights a dichotomy in the anti-association activity of eIF6 that is distinct from its role in 60S subunit biogenesis and shows that nucleolar localization of eIF6 is not dependent on BCCIP chaperone and uL14. Limiting active ribosomal pools markedly deregulates translation especially in mitosis and leads to chromosome segregation defects, mitotic exit delays and mitotic catastrophe. Ribo-seq analysis of eIF6-N106S mutant shows a significant downregulation in the translation efficiencies of mitotic factors and specifically transcripts with long 3' untranslated regions. eIF6-N106S mutation also limits cancer invasion, and this role is correlated with overexpression of eIF6 only in high-grade invasive cancers suggesting that deregulation of eIF6 is probably not an early event in cancers. Thus, this study highlights the segregation of eIF6 functions and its role in moderating 80S ribosome availability for translation, mitosis and cancer progression.
Collapse
Affiliation(s)
- Poonam Roshan
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Aparna Biswas
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Sinthyia Ahmed
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Stella Anagnos
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Riley Luebbers
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Kavya Harish
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Megan Li
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Nicholas Nguyen
- Division of Urologic Surgery, Saint Louis University School of Medicine, 6400 Clayton Road, Saint Louis, MO 63117, USA
| | - Gao Zhou
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Frank Tedeschi
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Vivian Hathuc
- Department of Pathology, Saint Louis University School of Medicine,1402 S. Grand Blvd., Saint Louis, MO 63104, USA
| | - Zhenguo Lin
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| | - Zachary Hamilton
- Division of Urologic Surgery, Saint Louis University School of Medicine, 6400 Clayton Road, Saint Louis, MO 63117, USA
| | - Sofia Origanti
- Department of Biology, Saint Louis University, 3507 Laclede Ave, Saint Louis, MO 63103, USA
| |
Collapse
|
2
|
Friedson B, Willis SD, Shcherbik N, Campbell AN, Cooper KF. The CDK8 kinase module: A novel player in the transcription of translation initiation and ribosomal genes. Mol Biol Cell 2025; 36:ar2. [PMID: 39565680 PMCID: PMC11742111 DOI: 10.1091/mbc.e24-04-0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
Survival following stress is dependent upon reprogramming transcription and translation. Communication between these programs following stress is critical for adaptation but is not clearly understood. The Cdk8 kinase module (CKM) of the Mediator complex modulates the transcriptional response to various stresses. Its involvement in regulating translational machinery has yet to be elucidated, highlighting an existing gap in knowledge. Here, we report that the CKM positively regulates a subset of ribosomal protein (RP) and translation initiation factor (TIF)-encoding genes under physiological conditions in Saccharomyces cerevisiae. In mouse embryonic fibroblasts and HCT116 cells, the CKM regulates unique sets of RP and TIF genes, demonstrating some conservation of function across species. In yeast, this is mediated by Cdk8 phosphorylation of one or more transcription factors which control RP and TIF expression. Conversely, the CKM is disassembled following nutrition stress, permitting repression of RP and TIF genes. The CKM also plays a transcriptional role important for promoting cell survival, particularly during translational machinery stress triggered by ribosome-targeting antibiotics. Furthermore, in mammalian cells, the activity of CDK8 and its paralogue, CDK19, promotes cell survival following ribosome inhibition. These results provide mechanistic insights into the CKM's role in regulating expression of a subset of genes associated with translation.
Collapse
Affiliation(s)
- Brittany Friedson
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Stephen D. Willis
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Natalia Shcherbik
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Alicia N. Campbell
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Katrina F. Cooper
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| |
Collapse
|
3
|
Isola JVV, Biswas S, Jayarathne H, Hubbart CR, Hense JD, Matsuzaki S, Kinter MT, Humphries KM, Ocañas SR, Sadagurski M, Stout MB. Canagliflozin treatment prevents follicular exhaustion and attenuates hallmarks of ovarian aging in genetically heterogenous mice. GeroScience 2024:10.1007/s11357-024-01465-w. [PMID: 39672978 DOI: 10.1007/s11357-024-01465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian aging is characterized by declines in follicular reserve and the emergence of mitochondrial dysfunction, reactive oxygen species production, inflammation, and fibrosis, which eventually results in menopause. Menopause is associated with increased systemic aging and the development of numerous comorbidities; therefore, the attenuation of ovarian aging could also delay systemic aging processes in women. Recent work has established that the anti-diabetic drug Canagliflozin (Cana), a sodium-glucose transporter 2 inhibitor, elicits benefits on aging-related outcomes, likely through the modulation of nutrient-sensing pathways and metabolic homeostasis. Given that nutrient-sensing pathways play a critical role in controlling primordial follicle activation, we sought to determine if chronic Cana administration would delay ovarian aging and curtail the emergence of pathological hallmarks associated with reproductive senescence. We found that mice receiving Cana maintained their ovarian reserve through 12 months of age, which was associated with declines in primordial follicles FoxO3a phosphorylation, a marker of activation, when compared to the age-matched controls. Furthermore, Cana treatment led to decreased collagen, lipofuscin, and T cell accumulation at 12 months of age. Whole ovary transcriptomic and proteomic analyses revealed subtle improvements, predominantly in mitochondrial function and the regulation of cellular proliferation. Pathway analyses of the transcriptomic data revealed a downregulation in cell proliferation and mitochondrial dysfunction signatures, with an upregulation of oxidative phosphorylation. Pathway analyses of the proteomic data revealed declines in signatures associated with PI3K/AKT activity and lymphocyte accumulation. Collectively, we demonstrate that Cana treatment can delay ovarian aging in mice and could potentially have efficacy for delaying ovarian aging in women.
Collapse
Affiliation(s)
- José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Subhasri Biswas
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Hashan Jayarathne
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Chase R Hubbart
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Jessica D Hense
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
4
|
Wang Z, Yang S, Tong L, Li X, Mao W, Yuan H, Chen Y, Zhang S, Zhang H, Chen R. eIF6 deficiency regulates gut microbiota, decreases systemic inflammation, and alleviates atherosclerosis. mSystems 2024; 9:e0059524. [PMID: 39225466 PMCID: PMC11494895 DOI: 10.1128/msystems.00595-24] [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: 05/11/2024] [Accepted: 07/20/2024] [Indexed: 09/04/2024] Open
Abstract
Altered composition of the gut microbiota affects immunity and metabolism. This study previously found that eIF6 gene knockdown changes the composition of the intestinal flora in the eIF6 gene knockdown mouse model. Lactobacillus acidophilus is significantly increased in the model. This study was designed to investigate the role of L. acidophilus in the pathogenesis of atherosclerosis. Transcriptomic data from 117 patients with coronary artery disease (CAD) and 79 healthy individuals were obtained. ApoE-/- and ApoE-/-/eIF6+/- mice on normal chow diet or a high-fat diet were treated for 16 weeks; eIF6 deficiency was evaluated atherosclerosis. ApoE-/- mice on normal chow diet or a high-fat diet were treated with L. acidophilus by daily oral gavage for 16 weeks. Moreover, one group was treated with lipopolysaccharide at 12 weeks. The levels of eIF6, RNASE3, and RSAD2 were notably higher in the patients with CAD than in the healthy individuals. eIF6 deficiency altered the composition of gut microbiota. eIF6 deficiency reduced the atherosclerotic lesion formation in ApoE-/-/eIF6+/- mice compared with the ApoE-/- mice. The microbial sequencing and metabolomics analysis demonstrated some beneficial bacterial (L. acidophilus, Ileibacterium, and Bifidobacterium) and metabolic levels significantly had deference in ApoE-/-/eIF6+/- mice compared with the ApoE-/- mice. Correlational studies indicated that L. acidophilus had close correlations with low-density lipoprotein cholesterol, lesion area, and necrotic area. L. acidophilus inhibited high-fat diet-induced inflammation and atherosclerotic lesion, increasing the expression of tight junction proteins (ZO-1 and claudin-1) and reducing the gut permeability. However, lipopolysaccharide reversed the protective effect of L. acidophilus against atherosclerosis. eIF6 deficiency protected against atherosclerosis by regulating the composition of gut microbiota and metabolites. L. acidophilus attenuated atherosclerotic lesions by reducing inflammation and increasing gut permeability.IMPORTANCEeIF6 deficiency modulates the gut microbiota and multiple metabolites in atherosclerotic ApoE-/- mice. L. acidophilus was reduced in the gut of atherosclerotic ApoE-/- mice, but administration of Lactobacillus acidophilus reversed intestinal barrier dysfunction and vascular inflammation. Our findings suggest that targeting individual species is a beneficial therapeutic strategy to prevent inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Shuai Yang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linglin Tong
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Li
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weiyi Mao
- School of Basic Medical Sciences, Nanjing Medical University, Jiangsu, China
| | - Honghua Yuan
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shenyang Zhang
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - He Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renjin Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
5
|
Sehrawat U. Exploiting Translation Machinery for Cancer Therapy: Translation Factors as Promising Targets. Int J Mol Sci 2024; 25:10835. [PMID: 39409166 PMCID: PMC11477148 DOI: 10.3390/ijms251910835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Eukaryotic protein translation has slowly gained the scientific community's attention for its advanced and powerful therapeutic potential. However, recent technical developments in studying ribosomes and global translation have revolutionized our understanding of this complex multistep process. These developments have improved and deepened the current knowledge of mRNA translation, sparking excitement and new possibilities in this field. Translation factors are crucial for maintaining protein synthesis homeostasis. Since actively proliferating cancer cells depend on protein synthesis, dysregulated protein translation is central to tumorigenesis. Translation factors and their abnormal expressions directly affect multiple oncogenes and tumor suppressors. Recently, small molecules have been used to target translation factors, resulting in translation inhibition in a gene-specific manner, opening the door for developing translation inhibitors that can lead to novel chemotherapeutic drugs for treating multiple cancer types caused by dysregulated translation machinery. This review comprehensively summarizes the involvement of translation factors in tumor progression and oncogenesis. Also, it sheds light on the evolution of translation factors as novel drug targets for developing future therapeutic drugs for treating cancer.
Collapse
Affiliation(s)
- Urmila Sehrawat
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
6
|
Biffo S, Ruggero D, Santoro MM. The crosstalk between metabolism and translation. Cell Metab 2024; 36:1945-1962. [PMID: 39232280 PMCID: PMC11586076 DOI: 10.1016/j.cmet.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
Metabolism and mRNA translation represent critical steps involved in modulating gene expression and cellular physiology. Being the most energy-consuming process in the cell, mRNA translation is strictly linked to cellular metabolism and in synchrony with it. Indeed, several mRNAs for metabolic pathways are regulated at the translational level, resulting in translation being a coordinator of metabolism. On the other hand, there is a growing appreciation for how metabolism impacts several aspects of RNA biology. For example, metabolic pathways and metabolites directly control the selectivity and efficiency of the translational machinery, as well as post-transcriptional modifications of RNA to fine-tune protein synthesis. Consistently, alterations in the intricate interplay between translational control and cellular metabolism have emerged as a critical axis underlying human diseases. A better understanding of such events will foresee innovative therapeutic strategies in human disease states.
Collapse
Affiliation(s)
- Stefano Biffo
- National Institute of Molecular Genetics and Biosciences Department, University of Milan, Milan, Italy.
| | - Davide Ruggero
- Department of Cellular and Molecular Pharmacology, Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Massimo Mattia Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
7
|
Huang CG, Zhou XQ, Zheng AF, Luo X, Shen J, Xiao ZG, Yang ZH, Dai Q. eIF6 Promotes Gastric Cancer Proliferation and Invasion by Regulating Cell Cycle. Dig Dis Sci 2024; 69:3249-3260. [PMID: 38987443 PMCID: PMC11415431 DOI: 10.1007/s10620-024-08464-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE To investigate the role and function of eIF6 in gastric cancer (GC). METHODS The expression level of eIF6 in GC tissues and normal tissues was detected in different high-throughput sequencing cohorts. Survival analysis, gene differential analysis, and enrichment analysis were performed in the TCGA cohort. Biological networks centered on eIF6 were constructed through two different databases. Immunohistochemistry (IHC) and Western blot were used to detect protein expression of eIF6, and qRT-PCR was used to detect eIF6 mRNA expression. The correlation between the expression of eIF6 in GC tissues and clinicopathological parameters of GC was analyzed. siRNA knockout of eIF6 was used to study the proliferation, migration, and invasion. The effects of eIF6 on cell cycle and Cyclin B1 were detected by flow cytometry and Western blot. RESULTS eIF6 was significantly overexpressed in GC tissues and predicted poor prognosis. In addition, 113 differentially expressed genes were detected in cancer-related biological pathways and functions by differential analysis. Biological networks revealed interactions of genes and proteins with eIF6. The expression intensity of eIF6 in cancer tissues was higher than that in adjacent tissues (P = 0.0001), confirming the up-regulation of eIF6 expression in GC tissues. The expression level of eIF6 was statistically significant with pTNM stage (P = 0.006). siRNA knockout of eIF6 significantly reduced the proliferation, colony formation, migration, and invasion ability of GC cells. Silencing of eIF6 also inhibited the cell cycle of GC cells in G2/M phase and decreased the expression level of CyclinB1. CONCLUSION Our study suggests that eIF6 is up-regulated in GC and may promote the proliferation, migration, and invasion of GC by regulating cell cycle.
Collapse
Affiliation(s)
- Cong-Gai Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Precision Pathology Diagnosis for Serious Diseases Key Laboratory of LuZhou, Luzhou, People's Republic of China
| | - Xiao-Qing Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - An-Fu Zheng
- Department of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Xing Luo
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jing Shen
- Department of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Zhan-Gang Xiao
- Department of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Zhi-Hui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Precision Pathology Diagnosis for Serious Diseases Key Laboratory of LuZhou, Luzhou, People's Republic of China
| | - Qiong Dai
- Department of Human Anatomy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
Roshan P, Biswas A, Anagnos S, Luebbers R, Harish K, Ahmed S, Li M, Nguyen N, Zhou G, Tedeschi F, Hathuc V, Lin Z, Hamilton Z, Origanti S. Modulation of ribosomal subunit associations by eIF6 is critical for mitotic exit and cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600220. [PMID: 38979253 PMCID: PMC11230244 DOI: 10.1101/2024.06.24.600220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Moderating the pool of active ribosomal subunits is critical for maintaining global translation rates. A factor crucial for modulating the 60S ribosomal subunits is eukaryotic translation initiation factor 6. Release of eIF6 from 60S is essential to permit 60S interactions with 40S. Here, using the N106S mutant of eIF6, we show that disrupting eIF6 interaction with 60S leads to an increase in vacant 80S. It further highlights a dichotomy in the anti-association activity of eIF6 that is distinct from its role in 60S biogenesis and shows that the nucleolar localization of eIF6 is not dependent on uL14-BCCIP interactions. Limiting active ribosomal pools markedly deregulates translation especially in mitosis and leads to chromosome segregation defects, mitotic exit delays and mitotic catastrophe. Ribo-Seq analysis of the eIF6-N106S mutant shows a significant downregulation in the translation efficiencies of mitotic factors and specifically transcripts with long 3'UTRs. eIF6-N106S mutation also limits cancer invasion, and this role is correlated with the overexpression of eIF6 only in high-grade invasive cancers suggesting that deregulation of eIF6 is probably not an early event in cancers. Thus, this study highlights the segregation of eIF6 functions and its role in moderating 80S availability for mitotic translation and cancer progression.
Collapse
|
9
|
Liu R, Cai R, Wang M, Zhang J, Zhang H, Li C, Sun C. Metagenomic insights into Heimdallarchaeia clades from the deep-sea cold seep and hydrothermal vent. ENVIRONMENTAL MICROBIOME 2024; 19:43. [PMID: 38909236 PMCID: PMC11193907 DOI: 10.1186/s40793-024-00585-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Heimdallarchaeia is a class of the Asgardarchaeota, are the most probable candidates for the archaeal protoeukaryote ancestor that have been identified to date. However, little is known about their life habits regardless of their ubiquitous distribution in diverse habitats, which is especially true for Heimdallarchaeia from deep-sea environments. In this study, we obtained 13 metagenome-assembled genomes (MAGs) of Heimdallarchaeia from the deep-sea cold seep and hydrothermal vent. These MAGs belonged to orders o_Heimdallarchaeales and o_JABLTI01, and most of them (9 MAGs) come from the family f_Heimdallarchaeaceae according to genome taxonomy database (GTDB). These are enriched for common eukaryote-specific signatures. Our results show that these Heimdallarchaeia have the metabolic potential to reduce sulfate (assimilatory) and nitrate (dissimilatory) to sulfide and ammonia, respectively, suggesting a previously unappreciated role in biogeochemical cycling. Furthermore, we find that they could perform both TCA and rTCA pathways coupled with pyruvate metabolism for energy conservation, fix CO2 and generate organic compounds through an atypical Wood-Ljungdahl pathway. In addition, many genes closely associated with bacteriochlorophyll and carotenoid biosynthesis, and oxygen-dependent metabolic pathways are identified in these Heimdallarchaeia MAGs, suggesting a potential light-utilization by pigments and microoxic lifestyle. Taken together, our results indicate that Heimdallarchaeia possess a mixotrophic lifestyle, which may give them more flexibility to adapt to the harsh deep-sea conditions.
Collapse
Affiliation(s)
- Rui Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Ruining Cai
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Minxiao Wang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Jing Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Huan Zhang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Chaolun Li
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.
| | - Chaomin Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.
| |
Collapse
|
10
|
Su Y, Shea J, DeStephanis D, Su Z. Transcriptomic Analysis of the Spatiotemporal Axis of Oogenesis and Fertilization in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597235. [PMID: 38895354 PMCID: PMC11185608 DOI: 10.1101/2024.06.03.597235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The oocyte germline of the C. elegans hermaphrodite presents a unique model to study the formation of oocytes. However, the size of the model animal and difficulties in retrieval of specific stages of the germline have obviated closer systematic studies of this process throughout the years. Here, we present a transcriptomic level analysis into the oogenesis of C. elegans hermaphrodites. We dissected a hermaphrodite gonad into seven sections corresponding to the mitotic distal region, the pachytene, the diplotene, the early diakinesis region and the 3 most proximal oocytes, and deeply sequenced the transcriptome of each of them along with that of the fertilized egg using a single-cell RNA-seq protocol. We identified specific gene expression events as well as gene splicing events in finer detail along the oocyte germline and provided novel insights into underlying mechanisms of the oogenesis process. Furthermore, through careful review of relevant research literature coupled with patterns observed in our analysis, we attempt to delineate transcripts that may serve functions in the interaction between the germline and cells of the somatic gonad. These results expand our knowledge of the transcriptomic space of the C. elegans germline and lay a foundation on which future studies of the germline can be based upon.
Collapse
Affiliation(s)
- Yangqi Su
- Department of Bioinformatics and Genomics, the University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Jonathan Shea
- Department of Bioinformatics and Genomics, the University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Darla DeStephanis
- Department of Bioinformatics and Genomics, the University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Zhengchang Su
- Department of Bioinformatics and Genomics, the University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
11
|
Taha MS, Ahmadian MR. Fragile X Messenger Ribonucleoprotein Protein and Its Multifunctionality: From Cytosol to Nucleolus and Back. Biomolecules 2024; 14:399. [PMID: 38672417 PMCID: PMC11047961 DOI: 10.3390/biom14040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene and a consequent lack of FMR protein (FMRP) synthesis are associated with fragile X syndrome, one of the most common inherited intellectual disabilities. FMRP is a multifunctional protein that is involved in many cellular functions in almost all subcellular compartments under both normal and cellular stress conditions in neuronal and non-neuronal cell types. This is achieved through its trafficking signals, nuclear localization signal (NLS), nuclear export signal (NES), and nucleolar localization signal (NoLS), as well as its RNA and protein binding domains, and it is modulated by various post-translational modifications such as phosphorylation, ubiquitination, sumoylation, and methylation. This review summarizes the recent advances in understanding the interaction networks of FMRP with a special focus on FMRP stress-related functions, including stress granule formation, mitochondrion and endoplasmic reticulum plasticity, ribosome biogenesis, cell cycle control, and DNA damage response.
Collapse
Affiliation(s)
- Mohamed S. Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
- Research on Children with Special Needs Department, Institute of Medical Research and Clinical Studies, National Research Centre, Cairo 12622, Egypt
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
12
|
Guo H, Lv J, Su X, Chen L, Ren J, Liu L, Ren M, Liu S, Dai M, Ren G, Gao F. Rice OseIF6.1 encodes a eukaryotic translation initiation factor and is essential for the development of grain and anther. FRONTIERS IN PLANT SCIENCE 2024; 15:1366986. [PMID: 38576779 PMCID: PMC10991840 DOI: 10.3389/fpls.2024.1366986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
The eIF6 proteins are distributed extensively in eukaryotes and play diverse and essential roles. The bona fide eIF6 protein in Arabidopsis, At-eIF6;1, is essential for embryogenesis. However, the role of eIF6 proteins in rice growth and development remains elusive and requires further investigation. Here, we characterized the functions of OseIF6.1, which is homologous to At-eIF6;1. OseIF6.1 encodes an eukaryotic translation initiation factor with a conserved eIF6 domain. The knockdown of OseIF6.1 resulted in a decrease in grain length and pollen sterility, whereas the overexpression of OseIF6.1 displayed opposite phenotypes. Further studies revealed that OseIF6.1 regulates grain shape by influencing cell expansion and proliferation. In addition, OseIF6.1 interacts with OsNMD3, which is a nuclear export adaptor for the 60S ribosomal subunit. The knockdown of OsNMD3 in plants exhibited reduced fertility and seed setting. Therefore, our findings have significantly enriched the current understanding of the role of OseIF6.1 in rice growth and development.
Collapse
Affiliation(s)
- Hongming Guo
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Jianqun Lv
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Xiangwen Su
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Liang Chen
- Xiamen Key Laboratory for Plant Genetics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Juansheng Ren
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Liping Liu
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Mingxin Ren
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Song Liu
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Mingli Dai
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Guangjun Ren
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Fangyuan Gao
- Environment-Friendly Crop Germplasm Innovation and Genetic Improvement Key Laboratory of Sichuan Province, Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
- Key Laboratory of Tianfu Seed Industry Innovation (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Chengdu, China
| |
Collapse
|
13
|
Key J, Gispert S, Kandi AR, Heinz D, Hamann A, Osiewacz HD, Meierhofer D, Auburger G. CLPP-Null Eukaryotes with Excess Heme Biosynthesis Show Reduced L-arginine Levels, Probably via CLPX-Mediated OAT Activation. Biomolecules 2024; 14:241. [PMID: 38397478 PMCID: PMC10886707 DOI: 10.3390/biom14020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
The serine peptidase CLPP is conserved among bacteria, chloroplasts, and mitochondria. In humans and mice, its loss causes Perrault syndrome, which presents with growth deficits, infertility, deafness, and ataxia. In the filamentous fungus Podospora anserina, CLPP loss leads to longevity. CLPP substrates are selected by CLPX, an AAA+ unfoldase. CLPX is known to target delta-aminolevulinic acid synthase (ALAS) to promote pyridoxal phosphate (PLP) binding. CLPX may also influence cofactor association with other enzymes. Here, the evaluation of P. anserina metabolomics highlighted a reduction in arginine/histidine levels. In Mus musculus cerebellum, reductions in arginine/histidine and citrulline occurred with a concomitant accumulation of the heme precursor protoporphyrin IX. This suggests that the increased biosynthesis of 5-carbon (C5) chain deltaALA consumes not only C4 succinyl-CoA and C1 glycine but also specific C5 delta amino acids. As enzymes responsible for these effects, the elevated abundance of CLPX and ALAS is paralleled by increased OAT (PLP-dependent, ornithine delta-aminotransferase) levels. Possibly as a consequence of altered C1 metabolism, the proteome profiles of P. anserina CLPP-null cells showed strong accumulation of a methyltransferase and two mitoribosomal large subunit factors. The reduced histidine levels may explain the previously observed metal interaction problems. As the main nitrogen-storing metabolite, a deficiency in arginine would affect the urea cycle and polyamine synthesis. Supplementation of arginine and histidine might rescue the growth deficits of CLPP-mutant patients.
Collapse
Affiliation(s)
- Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Suzana Gispert
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Arvind Reddy Kandi
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Daniela Heinz
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - Andrea Hamann
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - Heinz D. Osiewacz
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany;
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| |
Collapse
|
14
|
Chen Z, Wang F, Chen B, Wu G, Tian D, Yuan Q, Qiu S, Zhai Y, Chen J, Zheng H, Yan F. Turnip mosaic virus NIb weakens the function of eukaryotic translation initiation factor 6 facilitating viral infection in Nicotiana benthamiana. MOLECULAR PLANT PATHOLOGY 2024; 25:e13434. [PMID: 38388027 PMCID: PMC10883789 DOI: 10.1111/mpp.13434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/24/2024]
Abstract
Viruses rely completely on host translational machinery to produce the proteins encoded by their genes. Controlling translation initiation is important for gaining translational advantage in conflicts between the host and virus. The eukaryotic translation initiation factor 4E (eIF4E) has been reported to be hijacked by potyviruses for virus multiplication. The role of translation regulation in defence and anti-defence between plants and viruses is not well understood. We report that the transcript level of eIF6 was markedly increased in turnip mosaic virus (TuMV)-infected Nicotiana benthamiana. TuMV infection was impaired by overexpression of N. benthamiana eIF6 (NbeIF6) either transiently expressed in leaves or stably expressed in transgenic plants. Polysome profile assays showed that overexpression of NbeIF6 caused the accumulation of 40S and 60S ribosomal subunits, the reduction of polysomes, and also compromised TuMV UTR-mediated translation, indicating a defence role for upregulated NbeIF6 during TuMV infection. However, the polysome profile in TuMV-infected leaves was not identical to that in leaves overexpressing NbeIF6. Further analysis showed that TuMV NIb protein, the RNA-dependent RNA polymerase, interacted with NbeIF6 and interfered with its effect on the ribosomal subunits, suggesting that NIb might have a counterdefence role. The results propose a possible regulatory mechanism at the translation level during plant-virus interaction.
Collapse
Affiliation(s)
- Ziqiang Chen
- College of Life ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Biotechnology Research InstituteFujian Academy of Agricultural SciencesFuzhouChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Feng Wang
- Biotechnology Research InstituteFujian Academy of Agricultural SciencesFuzhouChina
| | - Binghua Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Guanwei Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Dagang Tian
- Biotechnology Research InstituteFujian Academy of Agricultural SciencesFuzhouChina
| | - Quan Yuan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Shiyou Qiu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Yushan Zhai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Jianping Chen
- College of Life ScienceFujian Agriculture and Forestry UniversityFuzhouChina
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Hongying Zheng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| | - Fei Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsInstitute of Plant Virology, Ningbo UniversityNingboChina
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang ProvinceInstitute of Plant Virology, Ningbo UniversityNingboChina
| |
Collapse
|
15
|
Xu R, Wang Y, Kuang Y. Multi-omic analyses of m5C readers reveal their characteristics and immunotherapeutic proficiency. Sci Rep 2024; 14:1651. [PMID: 38238581 PMCID: PMC10796763 DOI: 10.1038/s41598-024-52110-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/13/2024] [Indexed: 01/22/2024] Open
Abstract
5-methylcytosine (m5C) is a post-transcriptional RNA modification identified, m5C readers can specifically identify and bind to m5C. ALYREF and YBX1 as members of m5C readers that have garnered increasing attention in cancer research. However, comprehensive analysis of their molecular functions across pancancer are lacking. Using the TCGA and GTEx databases, we investigated the expression levels and prognostic values of ALYREF and YBX1. Additionally, we assessed the tumor microenvironment, immune checkpoint-related genes, immunomodulators, Tumor Immune Dysfunction and Exclusion (TIDE) score and drug resistance of ALYREF and YBX1. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) analyses were performed to investigate the potential functions associated with m5C readers and coexpressed genes. Aberrant expression of ALYREF and YBX1 was observed and positively associated with prognosis in KIRP, LGG and LIHC. Furthermore, the expression levels of ALYREF and YBX1 were significantly correlated with immune infiltration of the tumor microenvironment and immune-related modulators. Last, our analysis revealed significant correlations between ALYREF, YBX1 and eIFs. Our study provides a substantial understanding of m5C readers and the intricate relationship between ALYREF, YBX1, eIFs, and mRNA dynamics. Through multidimensional analysis of immune infiltration and drug sensitivity/resistance in ALYREF and YBX1, we propose a possibility for combined modality therapy utilizing m5C readers.
Collapse
Affiliation(s)
- Rui Xu
- Department of Development Planning, International Medical Opening-up Pilot Zone (China), Fangchenggang, Guangxi Province, China
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China.
| | - Ye Kuang
- Department of Medical Laboratory, Yan'An Hospital of Kunming City, Kunming, Yunnan Province, China.
| |
Collapse
|
16
|
Lao J, Sun H, Wang A, Wu M, Liu D, Zhang Y, Chen C, Xia Q, Ma S. Effect of eIF6 on the development of silk glands and silk protein synthesis of the silkworm, Bombyx mori. Int J Biol Macromol 2024; 256:128316. [PMID: 38000606 DOI: 10.1016/j.ijbiomac.2023.128316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023]
Abstract
The silkworm is a lepidopteran domesticated from the wild silkworm, mostly valued for its efficient synthesis of silk protein. This species' ability to spin silk has supported the 5500-year-old silk industry and the globally known "Silk Road", making the transformation of mulberry leaves into silk of great concern. Therefore, research on the silk-related genes of silkworms and their regulatory mechanisms has attracted increasing attention. Previous studies have revealed that domestic silk gland cells are endoreduplication cells, and their high-copy genome and special chromatin conformation provide conditions for the high expression of silk proteins. In this study, we systematically investigate the expression pattern of eukaryotic initiation factors (eIFs) and identified the eIF6 as a eukaryotic translation initiation factor involved in the synthesis of silk proteins. We generated an eIF6 gene deletion mutant strain of silkworm using the CRISPR/Cas9 system and investigated the function of eIF6 in silk gland development and silk protein synthesis. The results showed that deletion of eIF6 inhibited the individual development of silkworm larvae, inhibited the development of silk glands, and significantly reduced the cocoon layer ratio. Therefore, we elucidated the function of eIF6 in the development of silk glands and the synthesis of silk proteins, which is important for further elucidation of the developmental process of silk glands and the mechanism underlying the ultra-high expression of silk proteins.
Collapse
Affiliation(s)
- Junjie Lao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Hao Sun
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Aoming Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Mingke Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Dan Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Yan Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Chaojie Chen
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China; State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400716, China.
| | - Sanyuan Ma
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China; State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400716, China.
| |
Collapse
|
17
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
18
|
Toyoda K, Yasunaga JI, Shichijo T, Arima Y, Tsujita K, Tanaka A, Salah T, Zhang W, Hussein O, Sonoda M, Watanabe M, Kurita D, Nakashima K, Yamada K, Miyoshi H, Ohshima K, Matsuoka M. HTLV-1 bZIP Factor-Induced Reprogramming of Lactate Metabolism and Epigenetic Status Promote Leukemic Cell Expansion. Blood Cancer Discov 2023; 4:374-393. [PMID: 37162520 PMCID: PMC10473166 DOI: 10.1158/2643-3230.bcd-22-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/17/2023] [Accepted: 04/26/2023] [Indexed: 05/11/2023] Open
Abstract
Acceleration of glycolysis is a common trait of cancer. A key metabolite, lactate, is typically secreted from cancer cells because its accumulation is toxic. Here, we report that a viral oncogene, HTLV-1 bZIP factor (HBZ), bimodally upregulates TAp73 to promote lactate excretion from adult T-cell leukemia-lymphoma (ATL) cells. HBZ protein binds to EZH2 and reduces its occupancy of the TAp73 promoter. Meanwhile, HBZ RNA activates TAp73 transcription via the BATF3-IRF4 machinery. TAp73 upregulates the lactate transporters MCT1 and MCT4. Inactivation of TAp73 leads to intracellular accumulation of lactate, inducing cell death in ATL cells. Furthermore, TAp73 knockout diminishes the development of inflammation in HBZ-transgenic mice. An MCT1/4 inhibitor, syrosingopine, decreases the growth of ATL cells in vitro and in vivo. MCT1/4 expression is positively correlated with TAp73 in many cancers, and MCT1/4 upregulation is associated with dismal prognosis. Activation of the TAp73-MCT1/4 pathway could be a common mechanism contributing to cancer metabolism. SIGNIFICANCE An antisense gene encoded in HTLV-1, HBZ, reprograms lactate metabolism and epigenetic modification by inducing TAp73 in virus-positive leukemic cells. A positive correlation between TAp73 and its target genes is also observed in many other cancer cells, suggesting that this is a common mechanism for cellular oncogenesis. This article is featured in Selected Articles from This Issue, p. 337.
Collapse
Affiliation(s)
- Kosuke Toyoda
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun-ichirou Yasunaga
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takafumi Shichijo
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Azusa Tanaka
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tarig Salah
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Wenyi Zhang
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Osama Hussein
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Miyu Sonoda
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Miho Watanabe
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Kurita
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazutaka Nakashima
- Department of Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | - Kyohei Yamada
- Department of Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | - Masao Matsuoka
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Li C, Wang Y, Ge R, Zhang L, Du H, Zhang J, Li B, Chen K. Eukaryotic initiation factor 6 modulates the metamorphosis and reproduction of Tribolium castaneum. INSECT MOLECULAR BIOLOGY 2023; 32:106-117. [PMID: 36366777 DOI: 10.1111/imb.12817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Eukaryotic initiation factor 6 (eIF6) is necessary for ribosome biogenesis and translation, but eIF6 has been poorly elucidated in insects. Phylogenetic analysis demonstrated that eIF6 originated from one ancestral gene among animals and exhibited specific duplication in Tribolium, yielding three homologues in Tribolium castaneum, eIF6, eIF6-like 1 (eIF6l1), and eIF6-like 2 (eIF6l2). It was found that eIF6 was highly expressed in the embryonic and early adult stages, eIF6l1 had peak expression at the adult stage, and eIF6l2 showed peak expression in late adults of T. castaneum. Tissue-specific analyses in late-stage larvae demonstrated that eIF6 was abundantly expressed in all tissues, while eIF6l1 and eIF6l2 had the highest expression in the gut and the lowest expression in the head of T. castaneum. Knockdown of eIF6 caused precocious pupation and eclosion, impaired ovary and testis development and completely repressed egg production. The expression levels of vitellogenin 1 (Vg1), Vg2 and Vg receptor (VgR) significantly decreased in ds-eIF6 females 5 days post-adult emergence. Silencing eIF6 activated ecdysteroid biosynthesis and juvenile hormone degradation but reduced the activity of insulin signalling in T. castaneum, which might mediate its roles in metamorphosis, reproduction and gene expression regulation. However, silence of eIF6l1 or eIF6l2 had no effects on metamorphosis and reproduction in T. castaneum. This study provides comprehensive information for eIF6 evolution and function in the insect.
Collapse
Affiliation(s)
- Chengjun Li
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Youwei Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Runting Ge
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ling Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Huanyu Du
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Jiangyan Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
20
|
Translational Control of Metabolism and Cell Cycle Progression in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24054885. [PMID: 36902316 PMCID: PMC10002961 DOI: 10.3390/ijms24054885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The liver is a metabolic hub characterized by high levels of protein synthesis. Eukaryotic initiation factors, eIFs, control the first phase of translation, initiation. Initiation factors are essential for tumor progression and, since they regulate the translation of specific mRNAs downstream of oncogenic signaling cascades, may be druggable. In this review, we address the issue of whether the massive translational machinery of liver cells contributes to liver pathology and to the progression of hepatocellular carcinoma (HCC); it represents a valuable biomarker and druggable target. First, we observe that the common markers of HCC cells, such as phosphorylated ribosomal protein S6, belong to the ribosomal and translational apparatus. This fact is in agreement with observations that demonstrate a huge amplification of the ribosomal machinery during the progression to HCC. Some translation factors, such as eIF4E and eIF6, are then harnessed by oncogenic signaling. In particular, the action of eIF4E and eIF6 is particularly important in HCC when driven by fatty liver pathologies. Indeed, both eIF4E and eIF6 amplify at the translational level the production and accumulation of fatty acids. As it is evident that abnormal levels of these factors drive cancer, we discuss their therapeutic value.
Collapse
|
21
|
Elliff J, Biswas A, Roshan P, Kuppa S, Patterson A, Mattice J, Chinnaraj M, Burd R, Walker SE, Pozzi N, Antony E, Bothner B, Origanti S. Dynamic states of eIF6 and SDS variants modulate interactions with uL14 of the 60S ribosomal subunit. Nucleic Acids Res 2023; 51:1803-1822. [PMID: 36651285 PMCID: PMC9976893 DOI: 10.1093/nar/gkac1266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023] Open
Abstract
Assembly of ribosomal subunits into active ribosomal complexes is integral to protein synthesis. Release of eIF6 from the 60S ribosomal subunit primes 60S to associate with the 40S subunit and engage in translation. The dynamics of eIF6 interaction with the uL14 (RPL23) interface of 60S and its perturbation by somatic mutations acquired in Shwachman-Diamond Syndrome (SDS) is yet to be clearly understood. Here, by using a modified strategy to obtain high yields of recombinant human eIF6 we have uncovered the critical interface entailing eight key residues in the C-tail of uL14 that is essential for physical interactions between 60S and eIF6. Disruption of the complementary binding interface by conformational changes in eIF6 disease variants provide a mechanism for weakened interactions of variants with the 60S. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) analyses uncovered dynamic configurational rearrangements in eIF6 induced by binding to uL14 and exposed an allosteric interface regulated by the C-tail of eIF6. Disrupting key residues in the eIF6-60S binding interface markedly limits proliferation of cancer cells, which highlights the significance of therapeutically targeting this interface. Establishing these key interfaces thus provide a therapeutic framework for targeting eIF6 in cancers and SDS.
Collapse
Affiliation(s)
- Jonah Elliff
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
- Department of Immunology, The University of Iowa, Iowa City, IA 52242, USA
| | - Aparna Biswas
- Department of Biology, Saint Louis University, St. Louis, MO 63103, USA
| | - Poonam Roshan
- Department of Biology, Saint Louis University, St. Louis, MO 63103, USA
| | - Sahiti Kuppa
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, MO 63104, USA
| | - Angela Patterson
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Jenna Mattice
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Mathivanan Chinnaraj
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, MO 63104, USA
| | - Ryan Burd
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Sarah E Walker
- Department of Biological Sciences, State University of New York, Buffalo, NY 14260, USA
| | - Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, MO 63104, USA
| | - Edwin Antony
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, MO 63104, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Sofia Origanti
- Department of Biology, Saint Louis University, St. Louis, MO 63103, USA
| |
Collapse
|
22
|
Shen Y, Zhang R, Li X. Identification of eIF6 as a prognostic factor that drives tumor progression and predicts arsenic trioxide efficacy in lung adenocarcinoma. Mol Biol Rep 2023; 50:1167-1180. [PMID: 36435920 PMCID: PMC9889454 DOI: 10.1007/s11033-022-07917-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/03/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related mortality worldwide. Dysregulation of mRNA translation can contribute to the development and progression of cancer whilst also having an impact on the prognosis of different types of malignancies. Eukaryotic translation initiation factors (eIFs) have been reported to serve a key role in the initiation of mRNA translation. However, little was known about the association between eIF6 and lung adenocarcinoma (LUAD) progression. We aimed to elucidate the roles of eIF6 in LUAD tumorigenesis. METHODS Bioinformatic analysis was conducted to assess the clinical significance of eIF6 in LUAD. CCK-8, colony formation assays were used to evaluate the biological roles of eIF6. The subcutaneous model was used to assess the in vivo roles of eIF6. RESULTS In the present study, it was found that eIF6 expression was significantly higher in LUAD samples compared with that in normal lung tissues. Higher expression levels of eIF6 were found to be associated with more advanced clinical stages of LUAD and poorer prognoses in patients with LUAD. Subsequently, overexpression of eIF6 was demonstrated to promote LUAD cell proliferation, migration and invasion, which are features of metastasis, in vitro. By contrast, inhibition of eIF6 induced cell cycle arrest and apoptosis in LUAD cells. Further bioinformatics analysis and experimental assays revealed that eIF6 expression positively correlated with the mRNA expression of stemness-associated genes in LUAD cells. Targeting eIF6 suppressed the sphere formation capacity of LUAD cells. In addition, data from the subcutaneous xenograft model in vivo also suggested that eIF6 deficiency could significantly delay tumor growth and improve the prognosis of mice. Targeting eIF6 rendered LUAD cells sensitive to arsenic trioxide treatment. CONCLUSION The present study suggest that eIF6 can serve as a prognostic biomarker and a potential therapeutic target for patients with LUAD.
Collapse
Affiliation(s)
- Yan Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu P.R. China
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital, National Research Center for Translational Medicine at Shanghai, Shanghai Jiao Tong University School of Medicine, 200000 Shanghai, P.R. China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu P.R. China
| |
Collapse
|
23
|
Meng W, Xiao H, Mei P, Chen J, Wang Y, Zhao R, Liao Y. Critical Roles of METTL3 in Translation Regulation of Cancer. Biomolecules 2023; 13:biom13020243. [PMID: 36830614 PMCID: PMC9953158 DOI: 10.3390/biom13020243] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Aberrant translation, a characteristic feature of cancer, is regulated by the complex and sophisticated RNA binding proteins (RBPs) in the canonical translation machinery. N6-methyladenosine (m6A) modifications are the most abundant internal modifications in mRNAs mediated by methyltransferase-like 3 (METTL3). METTL3 is commonly aberrantly expressed in different tumors and affects the mRNA translation of many oncogenes or dysregulated tumor suppressor genes in a variety of ways. In this review, we discuss the critical roles of METTL3 in translation regulation and how METTL3 and m6A reader proteins in collaboration with RBPs within the canonical translation machinery promote aberrant translation in tumorigenesis, providing an overview of recent efforts aiming to 'translate' these results to the clinic.
Collapse
Affiliation(s)
- Wangyang Meng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Han Xiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiaping Chen
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence:
| |
Collapse
|
24
|
Chen Y, Wang Z, Xian X, Zhuang Y, Chang J, Zhan X, Han X, Chen Q, Yang Z, Chen R. Eukaryotic initiation factor 6 repression mitigates atherosclerosis progression by inhibiting macrophages expressing Fasn. IUBMB Life 2022; 75:440-452. [PMID: 36469534 DOI: 10.1002/iub.2696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Atherosclerosis, a chronic inflammatory disease that often leads to myocardial infarction and stroke, is mainly caused by lipid accumulation. Eukaryotic initiation factor 6 (Eif6) is a rate-limiting factor in protein translation of transcription factors necessary for lipogenesis. To determine whether Eif6 affects atherosclerosis, Eif6+/- mice were crossed into Apoe-/- background. Apoe-/-/Eif6+/- mice on high fat diet showed significant reduction in atherosclerotic lesions and necrotic core content in aortic root sections in comparison with Apoe-/- mice. RNA-Seq was used to investigate the effect of Eif6 in aorta. Deficiency of Eif6 shows broad effect on cell metabolism. Expression of genes for fatty acid synthesis including Fatty acid synthase (Fasn), Elovl3, Elovl6 and Acaca are down-regulated in aortas. Importantly, Fasn is decreased in macrophages. Results suggest that Eif6 deficiency may decrease atherosclerosis through inhibition of Fasn and lipids metabolism in macrophages.
Collapse
Affiliation(s)
- Yang Chen
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Zhenzhen Wang
- Cancer Institute Xuzhou Medical University Xuzhou China
| | - Xuemei Xian
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Yun Zhuang
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Jiajia Chang
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Xiaoqiang Zhan
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Xufeng Han
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Quangang Chen
- College of Life Sciences Xuzhou Medical University Xuzhou China
| | - Zhangping Yang
- Jiangsu Key Laboratory of Animal genetic Breeding and Molecular Design Yangzhou University Yangzhou China
| | - Renjin Chen
- College of Life Sciences Xuzhou Medical University Xuzhou China
| |
Collapse
|
25
|
Rubio A, Garland GD, Sfakianos A, Harvey RF, Willis AE. Aberrant protein synthesis and cancer development: The role of canonical eukaryotic initiation, elongation and termination factors in tumorigenesis. Semin Cancer Biol 2022; 86:151-165. [PMID: 35487398 DOI: 10.1016/j.semcancer.2022.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 01/27/2023]
Abstract
In tumourigenesis, oncogenes or dysregulated tumour suppressor genes alter the canonical translation machinery leading to a reprogramming of the translatome that, in turn, promotes the translation of selected mRNAs encoding proteins involved in proliferation and metastasis. It is therefore unsurprising that abnormal expression levels and activities of eukaryotic initiation factors (eIFs), elongation factors (eEFs) or termination factors (eRFs) are associated with poor outcome for patients with a wide range of cancers. In this review we discuss how RNA binding proteins (RBPs) within the canonical translation factor machinery are dysregulated in cancers and how targeting such proteins is leading to new therapeutic avenues.
Collapse
Affiliation(s)
- Angela Rubio
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Gavin D Garland
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Aristeidis Sfakianos
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Robert F Harvey
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK
| | - Anne E Willis
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge CB2 1QR, UK.
| |
Collapse
|
26
|
Huang C, Zhao Q, Zhou X, Huang R, Duan Y, Haybaeck J, Yang Z. The progress of protein synthesis factors eIFs, eEFs and eRFs in inflammatory bowel disease and colorectal cancer pathogenesis. Front Oncol 2022; 12:898966. [PMID: 36387239 PMCID: PMC9659945 DOI: 10.3389/fonc.2022.898966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/14/2022] [Indexed: 04/02/2025] Open
Abstract
Colorectal diseases are threatening human health, especially inflammatory bowel disease (IBD) and colorectal cancer (CRC). IBD is a group of chronic, recurrent and incurable disease, which may affect the entire gastrointestinal tract, increasing the risk of CRC. Eukaryotic gene expression is a complicated process, which is mainly regulated at the level of gene transcription and mRNA translation. Protein translation in tissue is associated with a sequence of steps, including initiation, elongation, termination and recycling. Abnormal regulation of gene expression is the key to the pathogenesis of CRC. In the early stages of cancer, it is vital to identify new diagnostic and therapeutic targets and biomarkers. This review presented current knowledge on aberrant expression of eIFs, eEFs and eRFs in colorectal diseases. The current findings of protein synthesis on colorectal pathogenesis showed that eIFs, eEFs and eRFs may be potential targets for CRC treatment.
Collapse
Affiliation(s)
- Conggai Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qi Zhao
- Department of Pathology, Basic Medical College of Southwest Medical University, Luzhou, China
| | - Xiaoqing Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ran Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Duan
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
27
|
Proteostasis Deregulation in Neurodegeneration and Its Link with Stress Granules: Focus on the Scaffold and Ribosomal Protein RACK1. Cells 2022; 11:cells11162590. [PMID: 36010666 PMCID: PMC9406587 DOI: 10.3390/cells11162590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
The role of protein misfolding, deposition, and clearance has been the dominant topic in the last decades of investigation in the field of neurodegeneration. The impairment of protein synthesis, along with RNA metabolism and RNA granules, however, are significantly emerging as novel potential targets for the comprehension of the molecular events leading to neuronal deficits. Indeed, defects in ribosome activity, ribosome stalling, and PQC—all ribosome-related processes required for proteostasis regulation—can contribute to triggering stress conditions and promoting the formation of stress granules (SGs) that could evolve in the formation of pathological granules, usually occurring during neurodegenerating effects. In this review, the interplay between proteostasis, mRNA metabolism, and SGs has been explored in a neurodegenerative context with a focus on Alzheimer’s disease (AD), although some defects in these same mechanisms can also be found in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), which are discussed here. Finally, we highlight the role of the receptor for activated C kinase 1 (RACK1) in these pathologies and note that, besides its well characterized function as a scaffold protein, it has an important role in translation and can associate to stress granules (SGs) determining cell fate in response to diverse stress stimuli.
Collapse
|
28
|
Liang J, Liu F, Yang Y, Li X, Cai G, Cao J, Zhang B. Diagnostic and prognostic utility of eIF6 in glioblastoma: a study based on TCGA and CGGA databases. Am J Transl Res 2022; 14:5040-5049. [PMID: 35958479 PMCID: PMC9360856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Among various glioma types, glioblastoma multiforme (GBM) is one of those with the highest malignancy. Although overexpression of eukaryotic translation initiation factor 6 (eIF6), a factor that regulates protein translation initiation, is believed to promote tumor development, its function and potential molecular mechanisms in glioma progression remain uncharacterized. Consequently, we evaluated its diagnostic and prognostic utility in GBM patients. METHODS Sample data from two databases, The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), were utilized to investigate the role of eIF6 as well as its mechanism of action in gliomas. We analyzed eIF6 expression in normal tissues as well as cancerous samples of different stages of glioma. The diagnostic and prognostic value of eIF6 were analyzed using the Receiver Operating Characteristic Curve (ROC) and Kaplan-Meier analysis, respectively. Furthermore, its underlying molecular mechanism in GBM was further revealed by gene set enrichment analysis (GSEA). RESULTS Transcriptome data analyses of the two databases showed that eIF6 was upregulated in glioma tissues compared with normal counterparts. eIF6 was at high levels in WHO grade IV gliomas versus grade II and III gliomas (P<0.05). In addition, eIF6 was highly expressed in elderly and Asian glioma patients. Furthermore, eIF6 expression was found to be lower in isocitrate dehydrogenase (IDH)-mutated tumors. Patients with high eIF6 level presented shorter overall survival than cases with low eIF6 level (P<0.05), and eIF6 had favorable accuracy in predicting the prognosis of glioma patients. GSEA revealed that high eIF6 expression was mainly concentrated in cell cycle and DNA repair related pathways. CONCLUSIONS eIF6 is highly expressed in gliomas and positively associated with the degree of malignancy. Patients with high eIF6 expression present poor survival. Therefore, eIF6 has the potential to be a diagnostic biomarker and a potential therapeutic target for glioma development and GBM.
Collapse
Affiliation(s)
- Jian Liang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Fengyu Liu
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Yaoqiang Yang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Xing Li
- School of Medicine, Southern University of Science and TechnologyShenzhen 518055, Guangdong, China
| | - Guangmou Cai
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Jianxuan Cao
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Bo Zhang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| |
Collapse
|
29
|
Wang Y, Qiao X, Li Y, Yang Q, Wang L, Liu X, Wang H, Shen H. Role of the receptor for activated C kinase 1 during viral infection. Arch Virol 2022; 167:1915-1924. [PMID: 35763066 DOI: 10.1007/s00705-022-05484-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/30/2022] [Indexed: 11/29/2022]
Abstract
Viruses can survive only in living cells, where they depend on the host's enzymatic system for survival and reproduction. Virus-host interactions are complex. On the one hand, hosts express host-restricted factors to protect the host cells from viral infections. On the other hand, viruses recruit certain host factors to facilitate their survival and transmission. The identification of host factors critical to viral infection is essential for comprehending the pathogenesis of contagion and developing novel antiviral therapies that specifically target the host. Receptor for activated C kinase 1 (RACK1), an evolutionarily conserved host factor that exists in various eukaryotic organisms, is a promising target for antiviral therapy. This review primarily summarizes the roles of RACK1 in regulating different viral life stages, particularly entry, replication, translation, and release.
Collapse
Affiliation(s)
- Yan Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiaorong Qiao
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yuhan Li
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Qingru Yang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Lulu Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiaolan Liu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hua Wang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hongxing Shen
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
30
|
Keen AN, Payne LA, Mehta V, Rice A, Simpson LJ, Pang KL, del Rio Hernandez A, Reader JS, Tzima E. Eukaryotic initiation factor 6 regulates mechanical responses in endothelial cells. J Cell Biol 2022; 221:e202005213. [PMID: 35024764 PMCID: PMC8763864 DOI: 10.1083/jcb.202005213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022] Open
Abstract
The repertoire of extratranslational functions of components of the protein synthesis apparatus is expanding to include control of key cell signaling networks. However, very little is known about noncanonical functions of members of the protein synthesis machinery in regulating cellular mechanics. We demonstrate that the eukaryotic initiation factor 6 (eIF6) modulates cellular mechanobiology. eIF6-depleted endothelial cells, under basal conditions, exhibit unchanged nascent protein synthesis, polysome profiles, and cytoskeleton protein expression, with minimal effects on ribosomal biogenesis. In contrast, using traction force and atomic force microscopy, we show that loss of eIF6 leads to reduced stiffness and force generation accompanied by cytoskeletal and focal adhesion defects. Mechanistically, we show that eIF6 is required for the correct spatial mechanoactivation of ERK1/2 via stabilization of an eIF6-RACK1-ERK1/2-FAK mechanocomplex, which is necessary for force-induced remodeling. These results reveal an extratranslational function for eIF6 and a novel paradigm for how mechanotransduction, the cellular cytoskeleton, and protein translation constituents are linked.
Collapse
Affiliation(s)
- Adam N. Keen
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Luke A. Payne
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Vedanta Mehta
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - Lisa J. Simpson
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kar Lai Pang
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Armando del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - John S. Reader
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellie Tzima
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Zhao Z, Chu W, Zheng Y, Wang C, Yang Y, Xu T, Yang X, Zhang W, Ding X, Li G, Zhang H, Zhou J, Ye J, Wu H, Song X, Wu Y. Cytoplasmic eIF6 promotes OSCC malignant behavior through AKT pathway. Cell Commun Signal 2021; 19:121. [PMID: 34922580 PMCID: PMC8684100 DOI: 10.1186/s12964-021-00800-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/30/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Eukaryotic translation initiation factor 6 (eIF6), also known as integrin β4 binding protein, is involved in ribosome formation and mRNA translation, acting as an anti-association factor. It is also essential for the growth and reproduction of cells, including tumor cells. Yet, its role in oral squamous cell carcinoma (OSCC) remains unclear. METHODS The expression characteristics of eIF6 in 233 samples were comprehensively analyzed by immunohistochemical staining (IHC). Effects of eIF6 over-expression and knockdown on cell proliferation, migration and invasion were determined by CCK-8, wound healing and Transwell assays. Western blot, immunofluorescence (IF) and co-immunoprecipitation (co-IP) were performed for mechanical verification. RESULTS We found that cytoplasmic eIF6 was abnormally highly expressed in OSCC tissues, and its expression was associated with tumor size and the clinical grade. Amplification of eIF6 promoted the growth, migration and invasion capabilities of OSCC cell lines in vitro and tumor growth in vivo. Through Western blot analysis, we further discovered that eIF6 significantly promotes epithelial-mesenchymal transformation (EMT) in OSCC cells, while depletion of eIF6 can reverse this process. Mechanistically, eIF6 promoted tumor progression by activating the AKT signaling pathway. By performing co-immunoprecipitation, we discovered a direct interaction between endogenous eIF6 and AKT protein in the cytoplasm. CONCLUSION These results demonstrated that eIF6 could be a new therapeutic target in OSCC, thus providing a new basis for the prognosis of OSCC patients in the future. Video abstract.
Collapse
Affiliation(s)
- Zechen Zhao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Weiming Chu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Stomatology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu People’s Republic of China
| | - Yang Zheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral Maxillofacial and Head and Neck Oncology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai, 200011 China
| | - Chao Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Yuemei Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Teng Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Xueming Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Stomatology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Wei Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Xu Ding
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Gang Li
- Department of Stomatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu People’s Republic of China
| | - Hongchuang Zhang
- Department of Stomatology, Xuzhou No.1 Peoples Hospital, Xuzhou, Jiangsu People’s Republic of China
| | - Junbo Zhou
- Department of Stomatology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, Jiangsu People’s Republic of China
| | - Jinhai Ye
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Heming Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Xiaomeng Song
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| | - Yunong Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No.1, Shanghai Road, Gulou District, Nanjing, Jiangsu 210029 People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu People’s Republic of China
| |
Collapse
|
32
|
Scagliola A, Miluzio A, Ventura G, Oliveto S, Cordiglieri C, Manfrini N, Cirino D, Ricciardi S, Valenti L, Baselli G, D'Ambrosio R, Maggioni M, Brina D, Bresciani A, Biffo S. Targeting of eIF6-driven translation induces a metabolic rewiring that reduces NAFLD and the consequent evolution to hepatocellular carcinoma. Nat Commun 2021; 12:4878. [PMID: 34385447 PMCID: PMC8361022 DOI: 10.1038/s41467-021-25195-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/24/2021] [Indexed: 12/30/2022] Open
Abstract
A postprandial increase of translation mediated by eukaryotic Initiation Factor 6 (eIF6) occurs in the liver. Its contribution to steatosis and disease is unknown. In this study we address whether eIF6-driven translation contributes to disease progression. eIF6 levels increase throughout the progression from Non-Alcoholic Fatty Liver Disease (NAFLD) to hepatocellular carcinoma. Reduction of eIF6 levels protects the liver from disease progression. eIF6 depletion blunts lipid accumulation, increases fatty acid oxidation (FAO) and reduces oncogenic transformation in vitro. In addition, eIF6 depletion delays the progression from NAFLD to hepatocellular carcinoma, in vivo. Mechanistically, eIF6 depletion reduces the translation of transcription factor C/EBPβ, leading to a drop in biomarkers associated with NAFLD progression to hepatocellular carcinoma and preserves mitochondrial respiration due to the maintenance of an alternative mTORC1-eIF4F translational branch that increases the expression of transcription factor YY1. We provide proof-of-concept that in vitro pharmacological inhibition of eIF6 activity recapitulates the protective effects of eIF6 depletion. We hypothesize the existence of a targetable, evolutionarily conserved translation circuit optimized for lipid accumulation and tumor progression.
Collapse
Affiliation(s)
- Alessandra Scagliola
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Annarita Miluzio
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | | | - Stefania Oliveto
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Chiara Cordiglieri
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Nicola Manfrini
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Delia Cirino
- Department of Biosciences, University of Milan, Milan, Italy
| | - Sara Ricciardi
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Guido Baselli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Roberta D'Ambrosio
- Department of Hepatology, Fondazione IRCCS Ca' Granda Granda Ospedale Policlinico, Milan, Italy
| | - Marco Maggioni
- Department of Pathology, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Daniela Brina
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alberto Bresciani
- Department of Translational and Discovery Research, IRBM S.p.A., Pomezia (Roma), Italy
| | - Stefano Biffo
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy.
- Department of Biosciences, University of Milan, Milan, Italy.
| |
Collapse
|
33
|
Sun L, Liu S, Wang X, Zheng X, Chen Y, Shen H. eIF6 promotes the malignant progression of human hepatocellular carcinoma via the mTOR signaling pathway. J Transl Med 2021; 19:216. [PMID: 34016142 PMCID: PMC8139032 DOI: 10.1186/s12967-021-02877-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Eukaryotic translation initiation factor 6 (eIF6) has a crucial function in the maturation of 60S ribosomal subunits, and it controls the initiation of protein translation. Although emerging studies indicate that eIF6 is aberrantly expressed in various types of cancers, the functions and underlying molecular mechanisms of eIF6 in the pathological progression of hepatocellular carcinoma (HCC) remain unclear. This study aimed to evaluate the potential diagnostic and prognostic value of eIF6 in patients with HCC. METHODS HCC samples enrolled from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and our cohort were used to explore the role and mechanism of eIF6 in HCC. The diagnostic power of eIF6 was verified by receiver operating characteristic curve (ROC) analysis and its prognostic value was assessed by Kaplan-Meier analysis, and then related biological functions of eIF6 were determined in vitro and in vivo cancer models. In addition, potential molecular mechanism of eIF6 in HCC was unveiled by the gene set enrichment analysis and western blot assay. RESULTS We demonstrated that eIF6 expression was markedly increased in HCC, and elevated eIF6 expression correlated with pathological progression of HCC. Besides, eIF6 served as not only a new diagnostic biomarker but also an independent risk factor for OS in HCC patients. Functional studies indicated that the deletion of eIF6 displayed tumor-suppressor activity in HCC cells. Furthermore, we found that eIF6 could activate the mTOR-related signaling pathway and regulate the expression level of its target genes, such as CCND1, CDK4, CDK6, MYC, CASP3 and CTNNBL1, and these activities promoted proliferation and invasion of HCC cells. CONCLUSIONS The findings of this study provided a novel basis for understanding the potential role of eIF6 in promoting tumor growth and invasion, and exploited a promising strategy for improving diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Liping Sun
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiaopai Wang
- Department of Pathology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Xuefeng Zheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Ya Chen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hong Shen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
34
|
Jungers CF, Elliff JM, Masson-Meyers DS, Phiel CJ, Origanti S. Regulation of eukaryotic translation initiation factor 6 dynamics through multisite phosphorylation by GSK3. J Biol Chem 2020; 295:12796-12813. [PMID: 32703900 DOI: 10.1074/jbc.ra120.013324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/16/2020] [Indexed: 01/25/2023] Open
Abstract
Eukaryotic translation initiation factor 6 (eIF6) is essential for the synthesis of 60S ribosomal subunits and for regulating the association of 60S and 40S subunits. A mechanistic understanding of how eIF6 modulates translation in response to stress, specifically starvation-induced stress, is lacking. We here show a novel mode of eIF6 regulation by glycogen synthase kinase 3 (GSK3) that is predominantly active in response to serum starvation. Both GSK3α and GSK3β phosphorylate human eIF6. Multiple residues in the C terminus of eIF6 are phosphorylated by GSK3 in a sequential manner. In response to serum starvation, eIF6 accumulates in the cytoplasm, and this altered localization depends on phosphorylation by GSK3. Disruption of eIF6 phosphorylation exacerbates the translation inhibitory response to serum starvation and stalls cell growth. These results suggest that eIF6 regulation by GSK3 contributes to the attenuation of global protein synthesis that is critical for adaptation to starvation-induced stress.
Collapse
Affiliation(s)
- Courtney F Jungers
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Jonah M Elliff
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | | | - Christopher J Phiel
- Department of Integrative Biology, University of Colorado Denver, Colorado, USA
| | - Sofia Origanti
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, USA .,Department of Biology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
35
|
Oyarbide U, Shah AN, Amaya-Mejia W, Snyderman M, Kell MJ, Allende DS, Calo E, Topczewski J, Corey SJ. Loss of Sbds in zebrafish leads to neutropenia and pancreas and liver atrophy. JCI Insight 2020; 5:134309. [PMID: 32759502 PMCID: PMC7526460 DOI: 10.1172/jci.insight.134309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 07/29/2020] [Indexed: 01/29/2023] Open
Abstract
Shwachman-Diamond syndrome (SDS) is characterized by exocrine pancreatic insufficiency, neutropenia, and skeletal abnormalities. Biallelic mutations in SBDS, which encodes a ribosome maturation factor, are found in 90% of SDS cases. Sbds–/– mice are embryonic lethal. Using CRISPR/Cas9 editing, we created sbds-deficient zebrafish strains. Sbds protein levels progressively decreased and became undetectable at 10 days postfertilization (dpf). Polysome analysis revealed decreased 80S ribosomes. Homozygous mutant fish developed normally until 15 dpf. Mutant fish subsequently had stunted growth and showed signs of atrophy in pancreas, liver, and intestine. In addition, neutropenia occurred by 5 dpf. Upregulation of tp53 mRNA did not occur until 10 dpf, and inhibition of proliferation correlated with death by 21 dpf. Transcriptome analysis showed tp53 activation through upregulation of genes involved in cell cycle arrest, cdkn1a and ccng1, and apoptosis, puma and mdm2. However, elimination of Tp53 function did not prevent lethality. Because of growth retardation and atrophy of intestinal epithelia, we studied the effects of starvation on WT fish. Starved WT fish showed intestinal atrophy, zymogen granule loss, and tp53 upregulation — similar to the mutant phenotype. In addition, there was reduction in neutral lipid storage and ribosomal protein amount, similar to the mutant phenotype. Thus, loss of Sbds in zebrafish phenocopies much of the human disease and is associated with growth arrest and tissue atrophy, particularly of the gastrointestinal system, at the larval stage. A variety of stress responses, some associated with Tp53, contribute to pathophysiology of SDS. Loss of ribosome maturation factor sbds in the zebrafish phenocopies human Shwachman-Diamond syndrome and is associated with p53 activation, but lethality cannot be rescued by p53 mutation.
Collapse
Affiliation(s)
- Usua Oyarbide
- Departments of Pediatrics, Immunology, and Human and Molecular Genetics, Children's Hospital of Richmond and Massey Cancer Center at Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Pediatrics, Stanley Manne Children's Research Institute, Northwestern University School of Medicine, Chicago, Illinois, USA.,Departments of Pediatrics, Cancer Biology, and Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Arish N Shah
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Wilmer Amaya-Mejia
- Departments of Pediatrics, Immunology, and Human and Molecular Genetics, Children's Hospital of Richmond and Massey Cancer Center at Virginia Commonwealth University, Richmond, Virginia, USA
| | - Matthew Snyderman
- Departments of Pediatrics, Cancer Biology, and Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Margaret J Kell
- Department of Pediatrics, Stanley Manne Children's Research Institute, Northwestern University School of Medicine, Chicago, Illinois, USA
| | | | - Eliezer Calo
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jacek Topczewski
- Department of Pediatrics, Stanley Manne Children's Research Institute, Northwestern University School of Medicine, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Seth J Corey
- Departments of Pediatrics, Immunology, and Human and Molecular Genetics, Children's Hospital of Richmond and Massey Cancer Center at Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Pediatrics, Stanley Manne Children's Research Institute, Northwestern University School of Medicine, Chicago, Illinois, USA.,Departments of Pediatrics, Cancer Biology, and Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
36
|
Manfrini N, Ricciardi S, Alfieri R, Ventura G, Calamita P, Favalli A, Biffo S. Ribosome profiling unveils translational regulation of metabolic enzymes in primary CD4 + Th1 cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 109:103697. [PMID: 32330465 DOI: 10.1016/j.dci.2020.103697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 05/22/2023]
Abstract
The transition from a naïve to an effector T cell is an essential event that requires metabolic reprogramming. We have recently demonstrated that the rapid metabolic changes that occur following stimulation of naïve T cells require the translation of preexisting mRNAs. Here, we provide evidence that translation regulates the metabolic asset of effector T cells. By performing ribosome profiling in human CD4+ Th1 cells, we show that the metabolism of glucose, fatty acids and pentose phosphates is regulated at the translational level. In Th1 cells, each pathway has at least one enzyme regulated at the translational level and selected enzymes have high translational efficiencies. mRNA expression does not predict protein expression. For instance, PKM2 mRNA is equally present in naïve T and Th1 cells, but the protein is abundant only in Th1. 5'-untranslated regions (UTRs) may partly account for this regulation. Overall we suggest that immunometabolism is controlled by translation.
Collapse
Affiliation(s)
- Nicola Manfrini
- INGM, National Institute of Molecular Genetics, "Fondazione Romeo ed Enrica Invernizzi", Milano, Italy; Department of Biological Sciences, University of Milan, Milan, Italy
| | - Sara Ricciardi
- INGM, National Institute of Molecular Genetics, "Fondazione Romeo ed Enrica Invernizzi", Milano, Italy; Department of Biological Sciences, University of Milan, Milan, Italy
| | - Roberta Alfieri
- INGM, National Institute of Molecular Genetics, "Fondazione Romeo ed Enrica Invernizzi", Milano, Italy
| | - Gabriele Ventura
- Department of Biological Sciences, University of Milan, Milan, Italy
| | - Piera Calamita
- INGM, National Institute of Molecular Genetics, "Fondazione Romeo ed Enrica Invernizzi", Milano, Italy; Department of Biological Sciences, University of Milan, Milan, Italy
| | - Andrea Favalli
- Department of Biological Sciences, University of Milan, Milan, Italy
| | - Stefano Biffo
- INGM, National Institute of Molecular Genetics, "Fondazione Romeo ed Enrica Invernizzi", Milano, Italy; Department of Biological Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
37
|
Impact of Eukaryotic Translation Initiation Factors on Breast Cancer: Still Much to Investigate. Cancers (Basel) 2020; 12:cancers12071984. [PMID: 32708122 PMCID: PMC7409344 DOI: 10.3390/cancers12071984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Breast carcinoma (BC) remains one of the most serious health problems. It is a heterogeneous entity, and mainly classified according to receptor status for estrogen (ER), progesterone (PR) and egf (HER2/Neu), as well as the proliferation marker ki67. Gene expression in eukaryotes is regulated at the level of both gene transcription and translation, where eukaryotic initiation factors (eIFs) are key regulators of protein biosynthesis. Aberrant translation results in an altered cellular proteome, and this clearly effects cell growth supporting tumorigenesis. The relationship between various eIFs and BC entities, as well as the related regulatory mechanisms, has meanwhile become a focus of scientific interest. Here, we give an overview on the current research state of eIF function, focusing on BC.
Collapse
|
38
|
Pesce E, Miluzio A, Turcano L, Minici C, Cirino D, Calamita P, Manfrini N, Oliveto S, Ricciardi S, Grifantini R, Degano M, Bresciani A, Biffo S. Discovery and Preliminary Characterization of Translational Modulators that Impair the Binding of eIF6 to 60S Ribosomal Subunits. Cells 2020; 9:cells9010172. [PMID: 31936702 PMCID: PMC7017188 DOI: 10.3390/cells9010172] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic initiation factor 6 (eIF6) is necessary for the nucleolar biogenesis of 60S ribosomes. However, most of eIF6 resides in the cytoplasm, where it acts as an initiation factor. eIF6 is necessary for maximal protein synthesis downstream of growth factor stimulation. eIF6 is an antiassociation factor that binds 60S subunits, in turn preventing premature 40S joining and thus the formation of inactive 80S subunits. It is widely thought that eIF6 antiassociation activity is critical for its function. Here, we exploited and improved our assay for eIF6 binding to ribosomes (iRIA) in order to screen for modulators of eIF6 binding to the 60S. Three compounds, eIFsixty-1 (clofazimine), eIFsixty-4, and eIFsixty-6 were identified and characterized. All three inhibit the binding of eIF6 to the 60S in the micromolar range. eIFsixty-4 robustly inhibits cell growth, whereas eIFsixty-1 and eIFsixty-6 might have dose- and cell-specific effects. Puromycin labeling shows that eIF6ixty-4 is a strong global translational inhibitor, whereas the other two are mild modulators. Polysome profiling and RT-qPCR show that all three inhibitors reduce the specific translation of well-known eIF6 targets. In contrast, none of them affect the nucleolar localization of eIF6. These data provide proof of principle that the generation of eIF6 translational modulators is feasible.
Collapse
Affiliation(s)
- Elisa Pesce
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
| | - Annarita Miluzio
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
| | - Lorenzo Turcano
- Department of Translational and Discovery Research, IRBM S.p.A., Via Pontina km 30, 600, 00071 Pomezia (Roma), Italy;
| | - Claudia Minici
- Biocrystallography Unit, Dept. of Immunology, Transplantation and Infectious Diseases, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132 Milan, Italy; (C.M.); (M.D.)
| | - Delia Cirino
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Piera Calamita
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Nicola Manfrini
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Stefania Oliveto
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Sara Ricciardi
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Renata Grifantini
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
| | - Massimo Degano
- Biocrystallography Unit, Dept. of Immunology, Transplantation and Infectious Diseases, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132 Milan, Italy; (C.M.); (M.D.)
| | - Alberto Bresciani
- Department of Translational and Discovery Research, IRBM S.p.A., Via Pontina km 30, 600, 00071 Pomezia (Roma), Italy;
- Correspondence: (A.B.); (S.B.)
| | - Stefano Biffo
- National Institute of Molecular Genetics, “Fondazione Romeo ed Enrica Invernizzi”, INGM, Via Francesco Sforza 35, 20122 Milan, Italy; (E.P.); (A.M.); (D.C.); (P.C.); (N.M.); (S.O.); (S.R.); (R.G.)
- DBS, University of Milan, Via Celoria 26, 20133 Milan, Italy
- Correspondence: (A.B.); (S.B.)
| |
Collapse
|
39
|
Kasari V, Pochopien AA, Margus T, Murina V, Turnbull K, Zhou Y, Nissan T, Graf M, Nováček J, Atkinson GC, Johansson MJO, Wilson DN, Hauryliuk V. A role for the Saccharomyces cerevisiae ABCF protein New1 in translation termination/recycling. Nucleic Acids Res 2019; 47:8807-8820. [PMID: 31299085 PMCID: PMC7145556 DOI: 10.1093/nar/gkz600] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/14/2019] [Accepted: 07/11/2019] [Indexed: 11/12/2022] Open
Abstract
Translation is controlled by numerous accessory proteins and translation factors. In the yeast Saccharomyces cerevisiae, translation elongation requires an essential elongation factor, the ABCF ATPase eEF3. A closely related protein, New1, is encoded by a non-essential gene with cold sensitivity and ribosome assembly defect knock-out phenotypes. Since the exact molecular function of New1 is unknown, it is unclear if the ribosome assembly defect is direct, i.e. New1 is a bona fide assembly factor, or indirect, for instance due to a defect in protein synthesis. To investigate this, we employed yeast genetics, cryo-electron microscopy (cryo-EM) and ribosome profiling (Ribo-Seq) to interrogate the molecular function of New1. Overexpression of New1 rescues the inviability of a yeast strain lacking the otherwise strictly essential translation factor eEF3. The structure of the ATPase-deficient (EQ2) New1 mutant locked on the 80S ribosome reveals that New1 binds analogously to the ribosome as eEF3. Finally, Ribo-Seq analysis revealed that loss of New1 leads to ribosome queuing upstream of 3′-terminal lysine and arginine codons, including those genes encoding proteins of the cytoplasmic translational machinery. Our results suggest that New1 is a translation factor that fine-tunes the efficiency of translation termination or ribosome recycling.
Collapse
Affiliation(s)
- Villu Kasari
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Building 6K and 6L, University Hospital Area, 90187 Umeå, Sweden
| | - Agnieszka A Pochopien
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Tõnu Margus
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Building 6K and 6L, University Hospital Area, 90187 Umeå, Sweden
| | - Victoriia Murina
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Building 6K and 6L, University Hospital Area, 90187 Umeå, Sweden
| | - Kathryn Turnbull
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Building 6K and 6L, University Hospital Area, 90187 Umeå, Sweden
| | - Yang Zhou
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden
| | - Tracy Nissan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, 10691, Sweden.,School of Life Science, University of Sussex, Brighton, BN19RH, UK
| | - Michael Graf
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Jiří Nováček
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Gemma C Atkinson
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden
| | - Marcus J O Johansson
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden
| | - Daniel N Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Vasili Hauryliuk
- Department of Molecular Biology, Umeå University, Building 6K, 6L University Hospital Area, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Building 6K and 6L, University Hospital Area, 90187 Umeå, Sweden.,University of Tartu, Institute of Technology, 50411 Tartu, Estonia
| |
Collapse
|
40
|
Golob-Schwarzl N, Wodlej C, Kleinegger F, Gogg-Kamerer M, Birkl-Toeglhofer AM, Petzold J, Aigelsreiter A, Thalhammer M, Park YN, Haybaeck J. Eukaryotic translation initiation factor 6 overexpression plays a major role in the translational control of gallbladder cancer. J Cancer Res Clin Oncol 2019; 145:2699-2711. [PMID: 31586263 PMCID: PMC6800842 DOI: 10.1007/s00432-019-03030-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gallbladder cancer (GBC) is a rare neoplasia of the biliary tract with high mortality rates and poor prognosis. Signs and symptoms of GBC are not specific and often arise at late stage of disease. For this reason, diagnosis is typically made when the cancer is already in advanced stages, and prognosis for survival is less than 5 years in 90% of cases. Biomarkers to monitor disease progression and novel therapeutic alternative targets for these tumors are strongly required. Commonly, dysregulated protein synthesis contributes to carcinogenesis and cancer progression. In this case, protein synthesis directs translation of specific mRNAs, and, in turn, promotes cell survival, invasion, angiogenesis, and metastasis of tumors. In eukaryotes, protein synthesis is regulated at its initiation, which is a rate-limiting step involving eukaryotic translation initiation factors (eIFs). We hypothesize that eIFs represent crossroads in the development of GBC, and might serve as potential biomarkers. The study focus was the role of eIF6 (an anti-association factor for the ribosomal subunits) in GBC. METHODS In human GBC samples, the expression of eIF6 was analyzed biochemically at the protein (immunohistochemistry, immunoblot analyses) and mRNA levels (qRT-PCR). RESULTS High levels of eIF6 correlated with shorter overall survival in biliary tract cancer (BTC) patients (n = 28). Immunohistochemical data from tissue microarrays (n = 114) demonstrated significantly higher expression levels of eIF6 in GBC compared to non-neoplastic tissue. Higher eIF6 expression on protein (immunoblot) and mRNA (qRT-PCR) level was confirmed by analyzing fresh frozen GBC patient samples (n = 14). Depletion of eIF6 (using specific siRNA-mediated knockdown) in Mz-ChA-2 and TFK-1 cell lines inhibited cell proliferation and induced apoptosis. CONCLUSION Our data indicates that eIF6 overexpression plays a major role in the translational control of GBC, and indicates its potential as a new biomarker and therapeutic target in GBC.
Collapse
Affiliation(s)
- Nicole Golob-Schwarzl
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Christina Wodlej
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine, Graz, Austria
| | - Florian Kleinegger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Department for Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Graz, Austria
| | - Margit Gogg-Kamerer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Johannes Petzold
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Ariane Aigelsreiter
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Michael Thalhammer
- Department of General Surgery, Medical University of Graz, Graz, Austria
| | - Young Nyun Park
- Department of Pathology, Yonsei University, College of Medicine Soul, Seoul, South Korea
| | - Johannes Haybaeck
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria.
- Center for Biomarker Research in Medicine, Graz, Austria.
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University, Leipziger Straße 44, 39210, Magdeburg, Germany.
- Department of Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
41
|
Modulating eIF6 levels unveils the role of translation in ecdysone biosynthesis during Drosophila development. Dev Biol 2019; 455:100-111. [DOI: 10.1016/j.ydbio.2019.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/01/2019] [Accepted: 05/28/2019] [Indexed: 11/18/2022]
|
42
|
US3 Kinase-Mediated Phosphorylation of Tegument Protein VP8 Plays a Critical Role in the Cellular Localization of VP8 and Its Effect on the Lipid Metabolism of Bovine Herpesvirus 1-Infected Cells. J Virol 2019; 93:JVI.02151-18. [PMID: 30626671 DOI: 10.1128/jvi.02151-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 02/04/2023] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) infects bovine species, causing respiratory infections, genital disorders and abortions. VP8 is the most abundant tegument protein of BoHV-1 and is critical for virus replication in cattle. In this study, the cellular transport of VP8 in BoHV-1-infected cells and its ability to alter the cellular lipid metabolism were investigated. A viral kinase, US3, was found to be involved in regulating these processes. In the early stages of infection VP8 was localized in the nucleus. Subsequently, presumably after completion of its role in the nucleus, VP8 was translocated to the cytoplasm. When US3 was deleted or the essential US3 phosphorylation site of VP8 was mutated in BoHV-1, the majority of VP8 was localized in the nuclei of infected cells. This suggests that phosphorylation by US3 may be critical for cytoplasmic localization of VP8. Eventually, the cytoplasmic VP8 was accumulated in the cis-Golgi apparatus but not in the trans-Golgi network, implying that VP8 was not involved in virion transport toward and budding from the cell membrane. VP8 caused lipid droplet (LD) formation in the nuclei of transfected cells and increased cellular cholesterol levels. Lipid droplets were not found in the nuclei of BoHV-1-infected cells when VP8 was cytoplasmic in the presence of US3. However, when US3 was deleted or phosphorylation residues in VP8 were mutated, nuclear VP8 and LDs appeared in BoHV-1-infected cells. The total cholesterol level was increased in BoHV-1-infected cells but not in ΔUL47-BoHV-1-infected cells, further supporting a role for VP8 in altering the cellular lipid metabolism during infection.IMPORTANCE Nuclear localization signals (NLSs) and nuclear export signals (NESs) are important elements directing VP8 to the desired locations in the BoHV-1-infected cell. In this study, a critical regulator that switches the nuclear and cytoplasmic localization of VP8 in BoHV-1-infected cells was identified. BoHV-1 used viral kinase US3 to regulate the cellular localization of VP8. Early during BoHV-1 infection VP8 was localized in the nucleus, where it performs various functions; once US3 was expressed, phosphorylated VP8 was cytoplasmic and ultimately accumulated in the cis-Golgi apparatus, presumably to be incorporated into virions. The Golgi localization of VP8 was only observed in virus-infected cells and not in US3-cotransfected cells, suggesting that this is mediated by other viral factors. Interestingly, VP8 was shown to cause increased cholesterol levels, which is a novel function for VP8 and a potential strategy to supply lipid for viral replication.
Collapse
|
43
|
Wang K, Wang C, Zhu CJ, Li G, Li Y, Feng YB, Ruan JJ, Zhu F, Meng Y, Zhou RP, Chen FH. 4-Amino-2-Trifluoromethyl-Phenyl Retinate induced leukemia cell differentiation by decreasing eIF6. Biochem Biophys Res Commun 2018; 503:2033-2039. [PMID: 30078681 DOI: 10.1016/j.bbrc.2018.07.153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 11/30/2022]
Abstract
4-Amino-2-Trifluoromethyl-Phenyl Retinate (ATPR), an all-trans retinoic acid (ATRA) derivative, possesses the ability to relief several carcinoma. Here, we explored the potential molecular mechanism of eukaryotic translation initiation factor 6 (eIF6) in ATPR-induced leukemia cell differentiation. Our research showed that ATPR could inhibit cell proliferation and promote cell differentiation in several leukemia cell lines. Besides, ATPR remarkably reduced the expression of eIF6 in vitro. Interestingly, the reduction of eIF6 contributed to restraining proliferation of K562 cells by inhibiting CyclinD1, C-myc and blocking cell cycle, as well as promoting differentiation of K562 cells by increasing the expression of C/EBPε, cell surface antigen CD11b and inducing renal-shrinkage of nuclear. Furthermore, the over-expression of eIF6 restrained the effects of ATPR on cell proliferation and maturation in K562 cells. In Addition, Notch1/CBF-1 signal activated by Chrysin could increase expression of eIF6 and restrain the differentiation in ATPR-induced K562 cells. Taken together, all above results indicated that ATPR induced differentiation of leukemia cells by decreasing eIF6 through Notch1/CBF-1 signal, which might exert an innovative treatment for leukemia.
Collapse
Affiliation(s)
- Ke Wang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Cong Wang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Chuan-Jun Zhu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Ge Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Yue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Yu-Bin Feng
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Jing-Jing Ruan
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Fei Zhu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Yao Meng
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Ren-Peng Zhou
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China
| | - Fei-Hu Chen
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, China.
| |
Collapse
|
44
|
Gaba A, Ayalew LE, Patel A, Kumar P, Tikoo SK. Bovine adenovirus‐3 protein VIII associates with eukaryotic initiation factor‐6 during infection. Cell Microbiol 2018. [DOI: 10.1111/cmi.12842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Amit Gaba
- VIDO‐InterVacUniversity of Saskatchewan Saskatoon Saskatchewan Canada
- Veterinary MicrobiologyUniversity of Saskatchewan Saskatoon Saskatchewan Canada
| | - Lisanework E. Ayalew
- VIDO‐InterVacUniversity of Saskatchewan Saskatoon Saskatchewan Canada
- Veterinary MicrobiologyUniversity of Saskatchewan Saskatoon Saskatchewan Canada
| | - Amrutlal Patel
- VIDO‐InterVacUniversity of Saskatchewan Saskatoon Saskatchewan Canada
- Veterinary MicrobiologyUniversity of Saskatchewan Saskatoon Saskatchewan Canada
| | - Pankaj Kumar
- VIDO‐InterVacUniversity of Saskatchewan Saskatoon Saskatchewan Canada
| | - Suresh K. Tikoo
- VIDO‐InterVacUniversity of Saskatchewan Saskatoon Saskatchewan Canada
- Veterinary MicrobiologyUniversity of Saskatchewan Saskatoon Saskatchewan Canada
- Vaccinology & Immunotherapeutics Program, School of Public HealthUniversity of Saskatchewan Saskatoon Saskatchewan Canada
| |
Collapse
|
45
|
Pfister AS, Kühl M. Of Wnts and Ribosomes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:131-155. [PMID: 29389514 DOI: 10.1016/bs.pmbts.2017.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Wnt proteins are secreted glycoproteins that activate different intracellular signal transduction pathways. They regulate cell proliferation and are required for proper embryonic development. Misregulation of Wnt signaling can result in various diseases including cancer. In most circumstances, cell growth is essential for cell division and thus cell proliferation. Therefore, several reports have highlighted the key role of Wnt proteins for cell growth. Ribosomes represent the cellular protein synthesis machinery and cells need to be equipped with an appropriate number of ribosomes to allow cell growth. Recent findings suggest a role for Wnt proteins in regulating ribosome biogenesis and we here summarize these findings representing a previously unknown function of Wnt proteins. Understanding this role of Wnt signaling might open new avenues to slow down proliferation by drugs for instance in cancer therapy.
Collapse
Affiliation(s)
- Astrid S Pfister
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany.
| | - Michael Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| |
Collapse
|
46
|
Manfrini N, Ricciardi S, Miluzio A, Fedeli M, Scagliola A, Gallo S, Brina D, Adler T, Busch DH, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Biffo S. High levels of eukaryotic Initiation Factor 6 (eIF6) are required for immune system homeostasis and for steering the glycolytic flux of TCR-stimulated CD4 + T cells in both mice and humans. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:69-76. [PMID: 28743432 DOI: 10.1016/j.dci.2017.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 06/07/2023]
Abstract
Eukaryotic Initiation Factor 6 (eIF6) is required for 60S ribosomal subunit biogenesis and efficient initiation of translation. Intriguingly, in both mice and humans, endogenous levels of eIF6 are detrimental as they act as tumor and obesity facilitators, raising the question on the evolutionary pressure that maintains high eIF6 levels. Here we show that, in mice and humans, high levels of eIF6 are required for proper immune functions. First, eIF6 heterozygous (het) mice show an increased mortality during viral infection and a reduction of peripheral blood CD4+ Effector Memory T cells. In human CD4+ T cells, eIF6 levels rapidly increase upon T-cell receptor activation and drive the glycolytic switch and the acquisition of effector functions. Importantly, in CD4+ T cells, eIF6 levels control interferon-γ (IFN-γ) secretion without affecting proliferation. In conclusion, the immune system has a high evolutionary pressure for the maintenance of a dynamic and powerful regulation of the translational machinery.
Collapse
Affiliation(s)
- Nicola Manfrini
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy.
| | - Sara Ricciardi
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy
| | - Annarita Miluzio
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy
| | - Maya Fedeli
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, DIBIT, H. San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Alessandra Scagliola
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy; Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Simone Gallo
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy; Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Daniela Brina
- Molecular Oncology Group, Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Via Mirasole 22A, Bellinzona, Switzerland
| | - Thure Adler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstrasse 30, 81675 Munich, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Stefano Biffo
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" - INGM, Via F. Sforza 35, 20122 Milan, Italy; Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy.
| |
Collapse
|
47
|
Kumar V, Fleming T, Terjung S, Gorzelanny C, Gebhardt C, Agrawal R, Mall MA, Ranzinger J, Zeier M, Madhusudhan T, Ranjan S, Isermann B, Liesz A, Deshpande D, Häring HU, Biswas SK, Reynolds PR, Hammes HP, Peperkok R, Angel P, Herzig S, Nawroth PP. Homeostatic nuclear RAGE-ATM interaction is essential for efficient DNA repair. Nucleic Acids Res 2017; 45:10595-10613. [PMID: 28977635 PMCID: PMC5737477 DOI: 10.1093/nar/gkx705] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022] Open
Abstract
The integrity of genome is a prerequisite for healthy life. Indeed, defects in DNA repair have been associated with several human diseases, including tissue-fibrosis, neurodegeneration and cancer. Despite decades of extensive research, the spatio-mechanical processes of double-strand break (DSB)-repair, especially the auxiliary factor(s) that can stimulate accurate and timely repair, have remained elusive. Here, we report an ATM-kinase dependent, unforeseen function of the nuclear isoform of the Receptor for Advanced Glycation End-products (nRAGE) in DSB-repair. RAGE is phosphorylated at Serine376 and Serine389 by the ATM kinase and is recruited to the site of DNA-DSBs via an early DNA damage response. nRAGE preferentially co-localized with the MRE11 nuclease subunit of the MRN complex and orchestrates its nucleolytic activity to the ATR kinase signaling. This promotes efficient RPA2S4-S8 and CHK1S345 phosphorylation and thereby prevents cellular senescence, IPF and carcinoma formation. Accordingly, loss of RAGE causatively linked to perpetual DSBs signaling, cellular senescence and fibrosis. Importantly, in a mouse model of idiopathic pulmonary fibrosis (RAGE−/−), reconstitution of RAGE efficiently restored DSB-repair and reversed pathological anomalies. Collectively, this study identifies nRAGE as a master regulator of DSB-repair, the absence of which orchestrates persistent DSB signaling to senescence, tissue-fibrosis and oncogenesis.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany
| | - Stefan Terjung
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany
| | - Christian Gorzelanny
- Experimental Dermatology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoffer Gebhardt
- Division of Dermatooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raman Agrawal
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, INF 156, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, INF 156, Heidelberg, Germany
| | - Julia Ranzinger
- Department of Nephrology, University of Heidelberg, Heidelberg, INF 410, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, INF 410, Heidelberg, Germany
| | - Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Satish Ranjan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD) University Hospital München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Divija Deshpande
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Subrata K Biswas
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, 3054 Life Sciences Building, Provo, UT 84602, USA
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rainer Peperkok
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Herzig
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany
| |
Collapse
|
48
|
Separation of low and high grade colon and rectum carcinoma by eukaryotic translation initiation factors 1, 5 and 6. Oncotarget 2017; 8:101224-101243. [PMID: 29254159 PMCID: PMC5731869 DOI: 10.18632/oncotarget.20642] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cause of cancer related death worldwide. Furthermore, with more than 1.2 million cases registered per year, it constitutes the third most frequent diagnosed cancer entity worldwide. Deregulation of protein synthesis has received considerable attention as a major step in cancer development and progression. Eukaryotic translation initiation factors (eIFs) are involved in the regulation of protein synthesis and are functionally linked to the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway. The identification of factors accounting for colorectal carcinoma (CRC) development is a major gap in the field. Besides the importance of eIF3 subunits and the eIF4 complex, eIF1, eIF5 and eIF6 were found to be altered in primary and metastatic CRC. We observed significant difference in the expression profile between low and high grade CRC. eIF1, eIF5 and eIF6 are involved in translational control in CRC. Our findings also indicate a probable clinical impact when separating them into low and high grade colon and rectum carcinoma. eIF and mTOR expression were analysed on protein and mRNA level in primary low and high grade colon carcinoma (CC) and rectum carcinoma (RC) samples in comparison to non-neoplastic tissue without any disease-related pathology. To assess the therapeutic potential of targeting eIF1, eIF5 and eIF6 siRNA knockdown in HCT116 and HT29 cells was performed. We evaluated the eIF knockdown efficacy on protein and mRNA level and investigated proliferation, apoptosis, invasion, as well as colony forming and polysome associated fractions. These results indicate that eIFs, in particular eIF1, eIF5 and eIF6 play a major role in translational control in colon and rectum cancer.
Collapse
|
49
|
Ali MU, Ur Rahman MS, Jia Z, Jiang C. Eukaryotic translation initiation factors and cancer. Tumour Biol 2017; 39:1010428317709805. [PMID: 28653885 DOI: 10.1177/1010428317709805] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent technological advancements have shown tremendous mechanistic accomplishments in our understanding of the mechanism of messenger RNA translation in eukaryotic cells. Eukaryotic messenger RNA translation is very complex process that includes four phases (initiation, elongation, termination, and ribosome recycling) and diverse mechanisms involving protein and non-protein molecules. Translation regulation is principally achieved during initiation step of translation, which is organized by multiple eukaryotic translation initiation factors. Eukaryotic translation initiation factor proteins help in stabilizing the formation of the functional ribosome around the start codon and provide regulatory mechanisms in translation initiation. Dysregulated messenger RNA translation is a common feature of tumorigenesis. Various oncogenic and tumor suppressive genes affect/are affected by the translation machinery, making the components of the translation apparatus promising therapeutic targets for the novel anticancer drug. This review provides details on the role of eukaryotic translation initiation factors in messenger RNA translation initiation, their contribution to onset and progression of tumor, and how dysregulated eukaryotic translation initiation factors can be used as a target to treat carcinogenesis.
Collapse
Affiliation(s)
- Muhammad Umar Ali
- 1 Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Muhammad Saif Ur Rahman
- 1 Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyu Jia
- 2 Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Cao Jiang
- 1 Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
50
|
Salton GD, Laurino CCFC, Mega NO, Delgado-Cañedo A, Setterblad N, Carmagnat M, Xavier RM, Cirne-Lima E, Lenz G, Henriques JAP, Laurino JP. Deletion of eIF2β lysine stretches creates a dominant negative that affects the translation and proliferation in human cell line: A tool for arresting the cell growth. Cancer Biol Ther 2017; 18:560-570. [PMID: 28692326 DOI: 10.1080/15384047.2017.1345383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Eukaryote initiation factor 2 subunit β (eIF2β) plays a crucial role in regulation protein synthesis, which mediates the interaction of eIF2 with mRNA. eIF2β contains evolutionarily conserved polylysine stretches in amino-terminal region and a zinc finger motif in the carboxy-terminus. METHODS The gene eIF2β was cloned under tetracycline transcription control and the polylysine stretches were deleted by site-directed mutagenesis (eIF2βΔ3K). The plasmid was transfected into HEK 293 TetR cells. These cells were analyzed for their proliferative and translation capacities as well as cell death rate. Experiments were performed using gene reporter assays, western blotting, flow cytometry, cell sorting, cell proliferation assays and confocal immunofluorescence. RESULTS eIF2βΔ3K affected negatively the protein synthesis, cell proliferation and cell survival causing G2 cell cycle arrest and increased cell death, acting in a negative dominant manner against the native protein. Polylysine stretches are also essential for eIF2β translocated from the cytoplasm to the nucleus, accumulating in the nucleolus and eIF2βΔ3K did not make this translocation. DISCUSSION eIF2β is involved in the protein synthesis process and should act in nuclear processes as well. eIF2βΔ3K reduces cell proliferation and causes cell death. Since translation control is essential for normal cell function and survival, the development of drugs or molecules that inhibit translation has become of great interest in the scenario of proliferative disorders. In conclusion, our results suggest the dominant negative eIF2βΔ3K as a therapeutic strategy for the treatment of proliferative disorders and that eIF2β polylysine stretch domains are promising targets for this.
Collapse
Affiliation(s)
- Gabrielle Dias Salton
- a Post-Graduation Program in Cellular and Molecular Biology, Molecular Radiobiology Laboratory, Biotechnology Center , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil , Cryobiology Unit and Umbilical Cord Blood Bank, Hemotherapy Service , Hospital de Clínicas de Porto Alegre , Porto Alegre (RS) , Brazil
| | - Claudia Cilene Fernandes Correia Laurino
- b Molecular Biology for Auto-immune and Infectious Diseases Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil . Embriology and Cellular Differentiation Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre; Faculdade de Veterinária , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil . Faculdade Nossa Senhora de Fátima , Caxias do Sul (RS) , Brazil . Instituto Brasileiro de Saúde , Porto Alegre (RS) , Brazil
| | - Nicolás Oliveira Mega
- c Animal Biology Post-Graduation Program , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil
| | - Andrés Delgado-Cañedo
- d Biotechnology Research Center for Interdisciplinary Research , Universidade Federal do Pampa , São Gabriel (RS) , Brazil
| | - Niclas Setterblad
- e Imaging, Cell Selection and Genomics Platform , Institut Universitaire d'Hématologie, Hôpital Saint-Louis , Paris , France
| | - Maryvonnick Carmagnat
- f Immunology and Histocompatibility Laboratory AP-HP , INSERM UMRS 940, Institut Universitaire d'Hématologie , Paris , France
| | - Ricardo Machado Xavier
- g Molecular Biology for Auto-immune and Infectious Diseases Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil
| | - Elizabeth Cirne-Lima
- h Embriology and Cellular Differentiation Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre; Faculdade de Veterinária , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil
| | - Guido Lenz
- i Cell Signaling Laboratory, Biophysics Department, Biotechnology Center and Post-Graduation Program in Cellular and Molecular Biology , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) , Brazil
| | - João Antonio Pêgas Henriques
- j Molecular Radiobiology Laboratory, Biotechnology Center and Post-Graduation Program in Cellular and Molecular Biology , Universidade Federal do Rio Grande do Sul , Porto Alegre (RS) ; Biotechnology Institute , Universidade de Caxias do Sul , Caxias do Sul (RS) , Brazil
| | - Jomar Pereira Laurino
- k Biotechnology Institute , Universidade de Caxias do Sul, Caxias do Sul (RS) and Instituto Brasileiro de Saúde , Porto Alegre (RS) , Brazil
| |
Collapse
|