1
|
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
|
2
|
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
|
3
|
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
|
4
|
Scagliola A, Miluzio A, Mori G, Ricciardi S, Oliveto S, Manfrini N, Biffo S. Inhibition of eIF6 Activity Reduces Hepatocellular Carcinoma Growth: An In Vivo and In Vitro Study. Int J Mol Sci 2022; 23:ijms23147720. [PMID: 35887068 PMCID: PMC9319760 DOI: 10.3390/ijms23147720] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by the accumulation of lipids in the liver. Given the high prevalence of NAFLD, its evolution to nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) is of global concern. Therapies for managing NASH-driven HCC can benefit from targeting factors that play a continuous role in NAFLD evolution to HCC. Recent work has shown that postprandial liver translation exacerbates lipid accumulation through the activity of a translation factor, eukaryotic initiation factor 6 (eIF6). Here, we test the effect of eIF6 inhibition on the progression of HCC. Mice heterozygous for eIF6 express half the level of eIF6 compared to wt mice and are resistant to the formation of HCC nodules upon exposure to a high fat/high sugar diet combined with liver damage. Histology showed that nodules in eIF6 het mice were smaller with reduced proliferation compared to wt nodules. By using an in vitro model of human HCC, we confirm that eIF6 depletion reduces the growth of HCC spheroids. We also tested three pharmacological inhibitors of eIF6 activity—eIFsixty-1, eIFsixty-4, and eIFsixty-6—and all three reduced eIF6 binding to 60S ribosomes and limited the growth of HCC spheroids. Thus, inhibition of eIF6 activity is feasible and limits HCC formation.
Collapse
Affiliation(s)
- Alessandra Scagliola
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Annarita Miluzio
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
| | - Giada Mori
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
| | - Sara Ricciardi
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Stefania Oliveto
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Nicola Manfrini
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
| | - Stefano Biffo
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, Via Sforza 35, 20122 Milan, Italy; (A.S.); (A.M.); (G.M.); (S.R.); (S.O.); (N.M.)
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy
- Correspondence:
| |
Collapse
|
5
|
Gao Y, Yuan L, Zeng J, Li F, Li X, Tan F, Liu X, Wan H, Kui X, Liu X, Ke C, Pei Z. eIF6 is potential diagnostic and prognostic biomarker that associated with 18F-FDG PET/CT features and immune signatures in esophageal carcinoma. Lab Invest 2022; 20:303. [PMID: 35794622 PMCID: PMC9258187 DOI: 10.1186/s12967-022-03503-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
Abstract
Background Although eukaryotic initiation factor 6 (eIF6) is a novel therapeutic target, data on its importance in the development of esophageal carcinoma (ESCA) remains limited. This study evaluated the correlation between eIF6 expression and metabolic analysis using fluorine-18 fluorodeoxyglucose (18F-FDG) -Positron emission tomography (PET) and immune gene signatures in ESCA. Methods This study employed The Cancer Genome Atlas (TCGA) to analyze the expression and prognostic value of eIF6, as well as its relationship with the immune gene signatures in ESCA patients. The qRT-PCR and Western blot analyses were used to profile the expression of eIF6 in ESCA tissues and different ESCA cell lines. The expression of tumor eIF6 and glucose transporter 1 (GLUT1) was examined using immunohistochemical tools in fifty-two ESCA patients undergoing routine 18F-FDG PET/CT before surgery. In addition, the cellular responses to eIF6 knockdown in human ESCA cells were assessed via the MTS, EdU, flow cytometry and wound healing assays. Results Our data demonstrated that compared with the normal esophageal tissues, eIF6 expression was upregulated in ESCA tumor tissues and showed a high diagnostic value with an area under curve of 0.825 for predicting ESCA. High eIF6 expression was significantly correlated with shorter overall survival of patients with esophagus adenocarcinoma (p = 0.038), but not in squamous cell carcinoma of the esophagus (p = 0.078). In addition, tumor eIF6 was significantly associated with 18F-FDG PET/CT parameters: maximal and mean standardized uptake values (SUVmax and SUVmean) and total lesion glycolysis (TLG) (rho = 0.458, 0.460, and 0.300, respectively, p < 0.01) as well as GLUT1 expression (rho = 0.453, p < 0.001). A SUVmax cutoff of 18.2 led to prediction of tumor eIF6 expression with an accuracy of 0.755. Functional analysis studies demonstrated that knockdown of eIF6 inhibited ESCA cell growth and migration, and fueled cell apoptosis. Moreover, the Bulk RNA gene analysis revealed a significant inverse association between eIF6 and the tumor-infiltrating immune cells (macrophages, T cells, or Th1 cells) and immunomodulators in the ESCA microenvironment. Conclusion Our study suggested that eIF6 might serve as a potential prognostic biomarker associated with metabolic variability and immune gene signatures in ESCA tumor microenvironment.
Collapse
|
6
|
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
|
7
|
Séité S, Harrison MC, Sillam-Dussès D, Lupoli R, Van Dooren TJM, Robert A, Poissonnier LA, Lemainque A, Renault D, Acket S, Andrieu M, Viscarra J, Sul HS, de Beer ZW, Bornberg-Bauer E, Vasseur-Cognet M. Lifespan prolonging mechanisms and insulin upregulation without fat accumulation in long-lived reproductives of a higher termite. Commun Biol 2022; 5:44. [PMID: 35027667 PMCID: PMC8758687 DOI: 10.1038/s42003-021-02974-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022] Open
Abstract
Kings and queens of eusocial termites can live for decades, while queens sustain a nearly maximal fertility. To investigate the molecular mechanisms underlying their long lifespan, we carried out transcriptomics, lipidomics and metabolomics in Macrotermes natalensis on sterile short-lived workers, long-lived kings and five stages spanning twenty years of adult queen maturation. Reproductives share gene expression differences from workers in agreement with a reduction of several aging-related processes, involving upregulation of DNA damage repair and mitochondrial functions. Anti-oxidant gene expression is downregulated, while peroxidability of membranes in queens decreases. Against expectations, we observed an upregulated gene expression in fat bodies of reproductives of several components of the IIS pathway, including an insulin-like peptide, Ilp9. This pattern does not lead to deleterious fat storage in physogastric queens, while simple sugars dominate in their hemolymph and large amounts of resources are allocated towards oogenesis. Our findings support the notion that all processes causing aging need to be addressed simultaneously in order to prevent it.
Collapse
Affiliation(s)
- Sarah Séité
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France
- University of Paris-Est, Créteil, France
| | - Mark C Harrison
- Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
| | - David Sillam-Dussès
- University Sorbonne Paris Nord, Laboratory of Experimental and Comparative Ethology, UR4443, Villetaneuse, France
| | - Roland Lupoli
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France
- University of Paris-Est, Créteil, France
| | - Tom J M Van Dooren
- UMR UPMC 113, IRD 242, UPEC, CNRS 7618, INRA 1392, PARIS 7 113, Institute of Ecology and Environmental Sciences of Paris, Paris, France
- Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Alain Robert
- University Sorbonne Paris Nord, Laboratory of Experimental and Comparative Ethology, UR4443, Villetaneuse, France
| | - Laure-Anne Poissonnier
- Department of Biochemistry, Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Arnaud Lemainque
- Genoscope, François-Jacob Institute of Biology, Alternative Energies and Atomic Energy Commission, University of Paris-Saclay, Evry, France
| | - David Renault
- University of Rennes, CNRS, ECOBIO (Ecosystems, biodiversity, evolution) - UMR, 6553, Rennes, France
- University Institute of France, Paris, France
| | - Sébastien Acket
- University of Technology of Compiègne, UPJV, UMR CNRS 7025, Enzyme and Cell Engineering, Royallieu research Center, Compiègne, France
| | - Muriel Andrieu
- Cochin Institute, UMR INSERM U1016, CNRS 8104, University of Paris Descartes, CYBIO Platform, Paris, France
| | - José Viscarra
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Z Wilhelm de Beer
- Department of Biochemistry, Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Erich Bornberg-Bauer
- Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
| | - Mireille Vasseur-Cognet
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France.
- University of Paris-Est, Créteil, France.
- INSERM, Paris, France.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Romano N, Catalani A, Lattante S, Belardo A, Proietti S, Bertini L, Silvestri F, Catalani E, Cervia D, Zolla L, Sabatelli M, Welshhans K, Ceci M. ALS skin fibroblasts reveal oxidative stress and ERK1/2-mediated cytoplasmic localization of TDP-43. Cell Signal 2020; 70:109591. [PMID: 32126264 DOI: 10.1016/j.cellsig.2020.109591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
The main hallmark of many forms of familiar and sporadic amyotrophic lateral sclerosis (ALS) is a reduction in nuclear TDP-43 protein and its inclusion in cytoplasmic aggregates in motor neurons. In order to understand which cellular and molecular mechanisms underlie the mislocalization of TDP-43, we examined human skin fibroblasts from two individuals with familial ALS, both with mutations in TDP-43, and two individuals with sporadic ALS, both without TDP-43 mutations or mutations in other ALS related genes. We found that all ALS fibroblasts had a partially cytoplasmic localization of TDP-43 and had reduced cell metabolism as compared to fibroblasts from apparently healthy individuals. ALS fibroblasts showed an increase in global protein synthesis and an increase in 4E-BP1 and rpS6 phosphorylation, which is indicative of mTORC1 activity. We also observed a decrease in glutathione (GSH), which suggests that oxidative stress is elevated in ALS. ERK1/2 activity regulated the extent of oxidative stress and the localization of TDP-43 in the cytoplasm in all ALS fibroblasts. Lastly, ALS fibroblasts showed reduced stress granule formation in response to H2O2 stress. In conclusion, these findings identify specific cellular and molecular defects in ALS fibroblasts, thus providing insight into potential mechanisms that may also occur in degenerating motor neurons.
Collapse
Affiliation(s)
- Nicla Romano
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Alessia Catalani
- Department of Molecular Sciences, University of Urbino "Carlo Bo", Via Santa Chiara, 27 61029 Urbino, PU, Italy
| | - Serena Lattante
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Unità Operativa Complessa di Genetica Medica, 00168 Roma, Italy; Università Cattolica del Sacro Cuore, Istituto di Medicina Genomica, 00168 Roma, Italy
| | - Antonio Belardo
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Silvia Proietti
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Laura Bertini
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Federica Silvestri
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Lello Zolla
- Department of Science and Technology for Agriculture, Forestry, Nature and Energy, University of Tuscia (DAFNE), 01100 Viterbo, Italy
| | - Mario Sabatelli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Neurologia, Dipartimento Scienze dell'invecchiamento, neurologiche, ortopediche e della testa-collo, 00168 Roma, Italy; Università Cattolica del Sacro Cuore, Istituto di Neurologia, Centro Clinico NEMO, 00168 Roma, Italy
| | - Kristy Welshhans
- Department of Biological Sciences, School of Biomedical Sciences and Brain Health Research Institute, Kent State University, Kent, OH 44236, USA
| | - Marcello Ceci
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy.
| |
Collapse
|
12
|
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
|
13
|
Drosophila RpS12 controls translation, growth, and cell competition through Xrp1. PLoS Genet 2019; 15:e1008513. [PMID: 31841522 PMCID: PMC6936874 DOI: 10.1371/journal.pgen.1008513] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 12/30/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Whereas complete loss of Rp function is generally lethal, most heterozygous Rp mutants grow more slowly and are subject to competitive loss from mosaics tissues that also contain wild type cells. The rpS12 gene has a special role in the cell competition of other Ribosomal Protein (Rp) mutant cells in Drosophila. Elimination by cell competition is promoted by higher RpS12 levels and prevented by a specific rpS12 mis-sense mutation, identifying RpS12 as a key effector of cell competition due to mutations in other Rp genes. Here we show that RpS12 is also required for other aspects of Rp mutant phenotypes, including hundreds of gene expression changes that occur in 'Minute' Rp heterozygous wing imaginal discs, overall translation rate, and the overall rate of organismal development, all through the bZip protein Xrp1 that is one of the RpS12-regulated genes. Our findings outline the regulatory response to mutations affecting essential Rp genes that controls overall translation, growth, and cell competition, and which may contribute to cancer and other diseases.
Collapse
|
14
|
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]
|
15
|
Robichaud N, Sonenberg N, Ruggero D, Schneider RJ. Translational Control in Cancer. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a032896. [PMID: 29959193 DOI: 10.1101/cshperspect.a032896] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The translation of messenger RNAs (mRNAs) into proteins is a key event in the regulation of gene expression. This is especially true in the cancer setting, as many oncogenes and transforming events are regulated at this level. Cancer-promoting factors that are translationally regulated include cyclins, antiapoptotic factors, proangiogenic factors, regulators of cell metabolism, prometastatic factors, immune modulators, and proteins involved in DNA repair. This review discusses the diverse means by which cancer cells deregulate and reprogram translation, and the resulting oncogenic impacts, providing insights into the complexity of translational control in cancer and its targeting for cancer therapy.
Collapse
Affiliation(s)
- Nathaniel Robichaud
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Nahum Sonenberg
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer Center, and Departments of Urology and of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158
| | - Robert J Schneider
- NYU School of Medicine, Alexandria Center for Life Science, New York, New York 10016
| |
Collapse
|
16
|
Somasundaram V, Basudhar D, Bharadwaj G, No JH, Ridnour LA, Cheng RY, Fujita M, Thomas DD, Anderson SK, McVicar DW, Wink DA. Molecular Mechanisms of Nitric Oxide in Cancer Progression, Signal Transduction, and Metabolism. Antioxid Redox Signal 2019; 30:1124-1143. [PMID: 29634348 PMCID: PMC6354612 DOI: 10.1089/ars.2018.7527] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/08/2018] [Indexed: 01/03/2023]
Abstract
SIGNIFICANCE Cancer is a complex disease, which not only involves the tumor but its microenvironment comprising different immune cells as well. Nitric oxide (NO) plays specific roles within tumor cells and the microenvironment and determines the rate of cancer progression, therapy efficacy, and patient prognosis. Recent Advances: Key understanding of the processes leading to dysregulated NO flux within the tumor microenvironment over the past decade has provided better understanding of the dichotomous role of NO in cancer and its importance in shaping the immune landscape. It is becoming increasingly evident that nitric oxide synthase 2 (NOS2)-mediated NO/reactive nitrogen oxide species (RNS) are heavily involved in cancer progression and metastasis in different types of tumor. More recent studies have found that NO from NOS2+ macrophages is required for cancer immunotherapy to be effective. CRITICAL ISSUES NO/RNS, unlike other molecules, are unique in their ability to target a plethora of oncogenic pathways during cancer progression. In this review, we subcategorize the different levels of NO produced by cells and shed light on the context-dependent temporal effects on cancer signaling and metabolic shift in the tumor microenvironment. FUTURE DIRECTIONS Understanding the source of NO and its spaciotemporal profile within the tumor microenvironment could help improve efficacy of cancer immunotherapies by improving tumor infiltration of immune cells for better tumor clearance.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Debashree Basudhar
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Gaurav Bharadwaj
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jae Hong No
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Lisa A. Ridnour
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Robert Y.S. Cheng
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Mayumi Fujita
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Department of Basic Medical Sciences for Radiation Damages, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Douglas D. Thomas
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Stephen K. Anderson
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Daniel W. McVicar
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - David A. Wink
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| |
Collapse
|
17
|
Kapadia B, Nanaji NM, Bhalla K, Bhandary B, Lapidus R, Beheshti A, Evens AM, Gartenhaus RB. Fatty Acid Synthase induced S6Kinase facilitates USP11-eIF4B complex formation for sustained oncogenic translation in DLBCL. Nat Commun 2018; 9:829. [PMID: 29483509 PMCID: PMC5827760 DOI: 10.1038/s41467-018-03028-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 01/15/2018] [Indexed: 01/29/2023] Open
Abstract
Altered lipid metabolism and aberrant protein translation are strongly associated with cancerous outgrowth; however, the inter-regulation of these key processes is still underexplored in diffuse large B-cell lymphoma (DLBCL). Although fatty acid synthase (FASN) activity is reported to positively correlate with PI3K-Akt-mTOR pathway that can modulate protein synthesis, the precise impact of FASN inhibition on this process is still unknown. Herein, we demonstrate that attenuating FASN expression or its activity significantly reduces eIF4B (eukaryotic initiation factor 4B) levels and consequently overall protein translation. Through biochemical studies, we identified eIF4B as a bonafide substrate of USP11, which stabilizes and enhances eIF4B activity. Employing both pharmacological and genetic approaches, we establish that FASN-induced PI3K-S6Kinase signaling phosphorylates USP11 enhancing its interaction with eIF4B and thereby promoting oncogenic translation.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Cell Line, Tumor
- Eukaryotic Initiation Factors/genetics
- Eukaryotic Initiation Factors/metabolism
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lipid Metabolism/genetics
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Protein Binding
- Protein Biosynthesis
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Ribosomal Protein S6 Kinases/genetics
- Ribosomal Protein S6 Kinases/metabolism
- Signal Transduction
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Thiolester Hydrolases/genetics
- Thiolester Hydrolases/metabolism
Collapse
Affiliation(s)
- Bandish Kapadia
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA
| | - Nahid M Nanaji
- Department of Veteran Affairs, Maryland Healthcare System, Baltimore, MD 21201, USA
- University of Maryland Medical Center, Baltimore, MD, 21201, USA
| | - Kavita Bhalla
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA
| | - Binny Bhandary
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA
| | - Rena Lapidus
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA
| | - Afshin Beheshti
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Andrew M Evens
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Ronald B Gartenhaus
- Department of Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA.
- Veterans Administration Medical Center, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
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
|
19
|
Manfrini N, Ricciardi S, Miluzio A, Fedeli M, Scagliola A, Gallo S, Adler T, Busch DH, Gailus-Durner V, Fuchs H, de Angelis MH, Biffo S. Data on the effects of eIF6 downmodulation on the proportions of innate and adaptive immune system cell subpopulations and on thymocyte maturation. Data Brief 2017; 14:653-658. [PMID: 28924581 PMCID: PMC5591389 DOI: 10.1016/j.dib.2017.08.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/02/2017] [Accepted: 08/24/2017] [Indexed: 11/28/2022] Open
Abstract
The data described in this article are related to “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” (Manfrini et al., in press) [1]. eIF6 is a translation initiation factor required for ribosomal biogenesis (Sanvito et al., 1999) [2] and for proper translational initiation (Gallo and Manfrini, 2015; Miluzio et al., 2016) [3], [4] whose protein abundance requires tight regulation. Here we analyze by flow cytometry the effects of eIF6 depletion on proportions of specific innate and adaptive immune system subpopulations and on thymocyte maturation in mice.
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
| | - 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
|