1
|
Tiburcio PD, Chen K, Xu L, Chen KS. Actinomycin D and bortezomib disrupt protein homeostasis in Wilms tumor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598518. [PMID: 38948702 PMCID: PMC11212905 DOI: 10.1101/2024.06.11.598518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Wilms tumor is the most common kidney cancer in children, and diffuse anaplastic Wilms tumor is the most chemoresistant histological subtype. Here, we explore how Wilms tumor cells evade the common chemotherapeutic drug actinomycin D, which inhibits ribosomal RNA biogenesis. Using ribosome profiling, protein arrays, and a genome-wide knockout screen, we describe how actinomycin D disrupts protein homeostasis and blocks cell cycle progression. We found that, when ribosomal capacity is limited by actinomycin D treatment, anaplastic Wilms tumor cells preferentially translate proteasome components and upregulate proteasome activity. Based on these findings, we tested whether the proteasome inhibitor bortezomib sensitizes cells to actinomycin D treatment. Indeed, we found that the combination induces apoptosis both in vitro and in vivo and prolongs survival in xenograft models. Lastly, we show that increased levels of proteasome components are associated with anaplastic histology and worse prognosis in Wilms tumor patients. In sum, maintaining protein homeostasis is critical for Wilms tumor proliferation, and it can be therapeutically disrupted by blocking protein synthesis or turnover.
Collapse
Affiliation(s)
| | - Kenian Chen
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX
| | - Lin Xu
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kenneth S. Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
2
|
Chatterjee S, Naeli P, Onar O, Simms N, Garzia A, Hackett A, Coyle K, Harris Snell P, McGirr T, Sawant TN, Dang K, Stoichkova Z, Azam Y, Saunders M, Braun M, Alain T, Tuschl T, McDade S, Longley D, Gkogkas C, Adrain C, Knight JP, Jafarnejad SM. Ribosome Quality Control mitigates the cytotoxicity of ribosome collisions induced by 5-Fluorouracil. Nucleic Acids Res 2024; 52:12534-12548. [PMID: 39351862 PMCID: PMC11551743 DOI: 10.1093/nar/gkae849] [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: 02/23/2024] [Revised: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 11/12/2024] Open
Abstract
Ribosome quality control (RQC) resolves collided ribosomes, thus preventing their cytotoxic effects. The chemotherapeutic agent 5-Fluorouracil (5FU) is best known for its misincorporation into DNA and inhibition of thymidylate synthase. However, while a major determinant of 5FU's anticancer activity is its misincorporation into RNAs, the mechanisms by which cancer cells overcome the RNA-dependent 5FU toxicity remain ill-defined. Here, we report a role for RQC in mitigating the cytotoxic effects of 5FU. We show that 5FU treatment results in rapid induction of the mTOR signalling pathway, enhanced rate of mRNA translation initiation, and increased ribosome collisions. Consistently, a defective RQC exacerbates the 5FU-induced cell death, which is mitigated by blocking mTOR pathway or mRNA translation initiation. Furthermore, 5FU treatment enhances the expression of the key RQC factors ZNF598 and GIGYF2 via an mTOR-dependent post-translational mechanism. This adaptation likely mitigates the cytotoxic consequences of increased ribosome collisions upon 5FU treatment.
Collapse
Affiliation(s)
- Susanta Chatterjee
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Parisa Naeli
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Okan Onar
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
- Department of Biology, Faculty of Science, Ankara University, Ankara, Turkey
| | - Nicole Simms
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Aitor Garzia
- Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Angela Hackett
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Kelsey Coyle
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Patric Harris Snell
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Tom McGirr
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Tanvi Nitin Sawant
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Kexin Dang
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Zornitsa Vasileva Stoichkova
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Yumna Azam
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Mark P Saunders
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Michael Braun
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ONK1H 8L1, Canada
| | - Thomas Tuschl
- Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Simon S McDade
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Daniel B Longley
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - Christos G Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Colin Adrain
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| | - John R P Knight
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9NT, UK
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT7 9AE, UK
| |
Collapse
|
3
|
Lee K, Poncz M. SLFN14 ribosomopathy and platelet dysfunction. Blood 2023; 141:2170-2172. [PMID: 37140956 PMCID: PMC10273156 DOI: 10.1182/blood.2023019949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Affiliation(s)
| | - Mortimer Poncz
- Children's Hospital of Philadelphia
- Perelman School of Medicine at the University of Pennsylvania
| |
Collapse
|
4
|
Ver Donck F, Ramaekers K, Thys C, Van Laer C, Peerlinck K, Van Geet C, Eto K, Labarque V, Freson K. Ribosome dysfunction underlies SLFN14-related thrombocytopenia. Blood 2023; 141:2261-2274. [PMID: 36790527 PMCID: PMC10646786 DOI: 10.1182/blood.2022017712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/16/2023] Open
Abstract
Pathogenic missense variants in SLFN14, which encode an RNA endoribonuclease protein that regulates ribosomal RNA (rRNA) degradation, are known to cause inherited thrombocytopenia (TP) with impaired platelet aggregation and adenosine triphosphate secretion. Despite mild laboratory defects, the patients displayed an obvious bleeding phenotype. However, the function of SLFN14 in megakaryocyte (MK) and platelet biology remains unknown. This study aimed to model the disease in an immortalized MK cell line (imMKCL) and to characterize the platelet transcriptome in patients with the SLFN14 K219N variant. MK derived from heterozygous and homozygous SLFN14 K219N imMKCL and stem cells of blood from patients mainly presented with a defect in proplatelet formation and mitochondrial organization. SLFN14-defective platelets and mature MK showed signs of rRNA degradation; however, this was absent in undifferentiated imMKCL cells and granulocytes. Total platelet RNA was sequenced in 2 patients and 19 healthy controls. Differential gene expression analysis yielded 2999 and 2888 significantly (|log2 fold change| >1, false discovery rate <0.05) up- and downregulated genes, respectively. Remarkably, these downregulated genes were not enriched in any biological pathway, whereas upregulated genes were enriched in pathways involved in (mitochondrial) translation and transcription, with a significant upregulation of 134 ribosomal protein genes (RPGs). The upregulation of mitochondrial RPGs through increased mammalian target of rapamycin complex 1 (mTORC1) signaling in SLFN14 K219N MK seems to be a compensatory response to rRNA degradation. mTORC1 inhibition with rapamycin resulted in further enhanced rRNA degradation in SLFN14 K219N MK. Taken together, our study indicates dysregulation of mTORC1 coordinated ribosomal biogenesis is the disease mechanism for SLFN14-related TP.
Collapse
Affiliation(s)
- Fabienne Ver Donck
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Kato Ramaekers
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Chantal Thys
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Christine Van Laer
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Clinical Department of Laboratory Medicine, Leuven University Hospitals, Leuven, Belgium
| | - Kathelijne Peerlinck
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Vascular Medicine and Hemostasis, Leuven University Hospitals, Leuven, Belgium
| | - Chris Van Geet
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Paediatric Hemato-Oncology, Leuven University Hospitals, Leuven, Belgium
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Veerle Labarque
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Paediatric Hemato-Oncology, Leuven University Hospitals, Leuven, Belgium
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Rozhkov SV, Sharlo KA, Shenkman BS, Mirzoev TM. Inhibition of mTORC1 differentially affects ribosome biogenesis in rat soleus muscle at the early and later stages of hindlimb unloading. Arch Biochem Biophys 2022; 730:109411. [PMID: 36155780 DOI: 10.1016/j.abb.2022.109411] [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: 06/21/2022] [Revised: 07/29/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
Prolonged inactivity of skeletal muscles due to limb immobilization, bedrest, and exposure to microgravity results in a significant muscle atrophy. Inactivity-induced muscle atrophy is caused by a downregulation of protein synthesis (PS) and increased proteolysis. Mechanistic target of rapamycin complex 1 (mTORC1) is considered to be one of the main regulators of translational capacity (quantity of ribosomes), a key determinant of PS. Using a specific mTORC1 inhibitor (rapamycin) we aimed to determine if mTORC1 activity would influence ribosome biogenesis in rat soleus muscle at both early and later stages of mechanical unloading. Wistar rats were subjected to 1- and 7-day hindlimb suspension (HS) with and without rapamycin injections (1.5 mg/kg) and compared to weight-bearing control animals. The key markers of ribosome biogenesis were assessed by RT-PCR or agarose gel electrophoresis. The rate of PS was measured by SUnSET method. Both 1-day and 7-day HS resulted in a significant downregulation of ribosome biogenesis markers (c-Myc, 47S pre-rRNA, 18S + 28S rRNAs) and the rate of PS. Rapamycin administration during 1-day HS fully prevented a decrease in 47S pre-rRNA expression and amount of 18S + 28S rRNAs (without affecting c-Myc mRNA expression) and partially attenuated a decline in PS. Rapamycin treatment during 7-day HS significantly decreased p70S6K phosphorylation but failed to rescue a reduction in both the markers of ribosome biogenesis and the rate of PS. All together, our results suggest that mTORC1 inhibition at the initial (1 day), but not later (7 days) stage of HS can be beneficial for the maintenance of translational capacity (ribosome biogenesis) and the rate of PS in rat soleus muscle.
Collapse
Affiliation(s)
- Sergey V Rozhkov
- Myology Laboratory, Institute of Biomedical Problems RAS, 123007, 76A Khoroshevskoe shosse, Moscow, Russia
| | - Kristina A Sharlo
- Myology Laboratory, Institute of Biomedical Problems RAS, 123007, 76A Khoroshevskoe shosse, Moscow, Russia
| | - Boris S Shenkman
- Myology Laboratory, Institute of Biomedical Problems RAS, 123007, 76A Khoroshevskoe shosse, Moscow, Russia
| | - Timur M Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, 123007, 76A Khoroshevskoe shosse, Moscow, Russia.
| |
Collapse
|
6
|
Small Nucleolar RNAs and Their Comprehensive Biological Functions in Hepatocellular Carcinoma. Cells 2022; 11:cells11172654. [PMID: 36078062 PMCID: PMC9454744 DOI: 10.3390/cells11172654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Small nucleolar RNAs (snoRNAs) are a class of highly conserved, stable non-coding RNAs involved in both post-transcriptional modification of RNA and in ribosome biogenesis. Recent research shows that the dysfunction of snoRNAs plays a pivotal role in hepatocellular carcinoma (HCC) and related etiologies, such as hepatitis B virus (HBV), hepatitis C virus (HCV), and non-alcoholic fatty liver disease (NAFLD). Growing evidence suggests that snoRNAs act as oncogenes or tumor suppressors in hepatocellular carcinoma (HCC) through multiple mechanisms. Furthermore, snoRNAs are characterized by their stability in body fluids and their clinical relevance and represent promising tools as diagnostic and prognostic biomarkers. SnoRNAs represent an emerging area of cancer research. In this review, we summarize the classification, biogenesis, activity, and functions of snoRNAs, as well as highlight the mechanism and roles of snoRNAs in HCC and related diseases. Our findings will aid in the understanding of complex processes of tumor occurrence and development, as well as suggest potential diagnostic markers and treatment targets. Furthermore, we discuss several limitations and suggest future research and application directions.
Collapse
|
7
|
Functions of block of proliferation 1 during anterior development in Xenopus laevis. PLoS One 2022; 17:e0273507. [PMID: 36007075 PMCID: PMC9409556 DOI: 10.1371/journal.pone.0273507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022] Open
Abstract
Block of proliferation 1 (Bop1) is a nucleolar protein known to be necessary for the assembly of the 60S subunit of ribosomes. Here, we show a specific bop1 expression in the developing anterior tissue of the South African clawed frog Xenopus laevis. Morpholino oligonucleotide-mediated knockdown approaches demonstrated that Bop1 is required for proper development of the cranial cartilage, brain, and the eyes. Furthermore, we show that bop1 knockdown leads to impaired retinal lamination with disorganized cell layers. Expression of neural crest-, brain-, and eye-specific marker genes was disturbed. Apoptotic and proliferative processes, which are known to be affected during ribosomal biogenesis defects, are not hindered upon bop1 knockdown. Because early Xenopus embryos contain a large store of maternal ribosomes, we considered if Bop1 might have a role independent of de novo ribosomal biogenesis. At early embryonic stages, pax6 expression was strongly reduced in bop1 morphants and synergy experiments indicate a common signaling pathway of the two molecules, Bop1 and Pax6. Our studies imply a novel function of Bop1 independent of ribosomal biogenesis.
Collapse
|
8
|
Abdelrahman SA, Abdelfatah MM, Keshta AT. Rapamycin-filgrastim combination therapy ameliorates portal hypertension-induced splenomegaly: Role of β actin and S100A9 proteins modulation. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:732-744. [PMID: 35949314 PMCID: PMC9320204 DOI: 10.22038/ijbms.2022.64034.14101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/07/2022] [Indexed: 11/06/2022]
Abstract
Objectives Thioacetamide (TAA) was administered to induce an animal model of liver disease with secondary splenomegaly to assess the mechanisms underlying the effects of rapamycin and filgrastim when taken separately or in combination on the biochemical and histopathological aspects of the liver and spleen. Materials and Methods Thirty adult male albino rats were divided into five groups (control, TAA-treated group, TAA+rapamycin, TAA+filgrastim, and TAA+rapamycin+filgrastim group). We measured relative liver and spleen weights, serum levels of alanine transaminase (ALT), aspartate transaminase (AST), and albumin. Molecular docking modeling and histopathological examination of liver and spleen sections with hematoxylin and eosin and Masson trichrome staining with immunohistochemical detection of splenic CD3 and CD20 lymphocytes, S100A9 and β actin antibodies were detected. Morphometric and statistical analyses of the results were performed. Results TAA administration altered the histological structure of the liver and spleen and impaired liver function. It increased the expression of splenic CD3, CD20 lymphocytes, and S100A9 while diminishing the expression of β actin. Each of rapamycin and filgrastim, when administered separately, improved liver and spleen indices and liver function, but rapamycin did not affect the albumin level. They lowered splenic B and T lymphocyte levels. Expression levels of S100A9 showed down-regulation while β actin levels were up-regulated when compared with TAA. Combination therapy improved liver and spleen tissue pathology and significantly ameliorated the expression of splenic lymphocytes through regulation of S100A9 and β actin expression. Conclusion The synergistic effect of combination therapy was dependent on the regulation of splenic S100A9 and β actin levels.
Collapse
Affiliation(s)
- Shaimaa A. Abdelrahman
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt,Corresponding author: Shaimaa A. Abdelrahman. Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Mohammed M. Abdelfatah
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Akaber T. Keshta
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
9
|
BOP1 Used as a Novel Prognostic Marker and Correlated with Tumor Microenvironment in Pan-Cancer. JOURNAL OF ONCOLOGY 2021; 2021:3603030. [PMID: 34603446 PMCID: PMC8481050 DOI: 10.1155/2021/3603030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/24/2021] [Accepted: 09/04/2021] [Indexed: 01/18/2023]
Abstract
Previous studies have indicated the important role of block of proliferation 1 (BOP1) in the progression of several malignant tumors; no comprehensive pan-cancer analysis of BOP1 has been performed. Here, we aim to systematically identify the expression, prognostic value, and potential immunological functions of BOP1 in 33 malignancies. We obtained the gene expression data and clinical information from multiple public databases to assess the expression level and prognostic value of BOP1 in 33 cancers. We also analyzed the relationship between BOP1 expression and DNA methylation, tumor microenvironment (TME), microsatellite instability (MSI), tumor mutational burden (TMB), and immune checkpoints. Moreover, we conducted gene set enrichment analysis (GSEA) to investigate the biological function and signal transduction pathways of BOP1 in different types of tumors. Finally, we validated the expression of BOP1 in lung cancer cell line and detected the influence of BOP1 on lung cancer cell migration and the expression of epithelial-mesenchymal transition- (EMT-) related genes. Collectively, our findings elucidated that BOP1 has the potential to be a promising molecular prognostic biomarker for predicting poor survival in various malignant tumors, as well as a cancer-promoting gene involved in tumorigenesis and tumor immunity.
Collapse
|
10
|
Sletten AC, Davidson JW, Yagabasan B, Moores S, Schwaiger-Haber M, Fujiwara H, Gale S, Jiang X, Sidhu R, Gelman SJ, Zhao S, Patti GJ, Ory DS, Schaffer JE. Loss of SNORA73 reprograms cellular metabolism and protects against steatohepatitis. Nat Commun 2021; 12:5214. [PMID: 34471131 PMCID: PMC8410784 DOI: 10.1038/s41467-021-25457-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia and resulting lipotoxicity are pathologic signatures of metabolic syndrome and type 2 diabetes. Excess lipid causes cell dysfunction and induces cell death through pleiotropic mechanisms that link to oxidative stress. However, pathways that regulate the response to metabolic stress are not well understood. Herein, we show that disruption of the box H/ACA SNORA73 small nucleolar RNAs encoded within the small nucleolar RNA hosting gene 3 (Snhg3) causes resistance to lipid-induced cell death and general oxidative stress in cultured cells. This protection from metabolic stress is associated with broad reprogramming of oxidative metabolism that is dependent on the mammalian target of rapamycin signaling axis. Furthermore, we show that knockdown of SNORA73 in vivo protects against hepatic steatosis and lipid-induced oxidative stress and inflammation. Our findings demonstrate a role for SNORA73 in the regulation of metabolism and lipotoxicity.
Collapse
Affiliation(s)
- Arthur C Sletten
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Busra Yagabasan
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Samantha Moores
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Hideji Fujiwara
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah Gale
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Xuntian Jiang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Rohini Sidhu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Susan J Gelman
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Shuang Zhao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel S Ory
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jean E Schaffer
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Ballard DJ, Peng HY, Das JK, Kumar A, Wang L, Ren Y, Xiong X, Ren X, Yang JM, Song J. Insights Into the Pathologic Roles and Regulation of Eukaryotic Elongation Factor-2 Kinase. Front Mol Biosci 2021; 8:727863. [PMID: 34532346 PMCID: PMC8438118 DOI: 10.3389/fmolb.2021.727863] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic Elongation Factor-2 Kinase (eEF2K) acts as a negative regulator of protein synthesis, translation, and cell growth. As a structurally unique member of the alpha-kinase family, eEF2K is essential to cell survival under stressful conditions, as it contributes to both cell viability and proliferation. Known as the modulator of the global rate of protein translation, eEF2K inhibits eEF2 (eukaryotic Elongation Factor 2) and decreases translation elongation when active. eEF2K is regulated by various mechanisms, including phosphorylation through residues and autophosphorylation. Specifically, this protein kinase is downregulated through the phosphorylation of multiple sites via mTOR signaling and upregulated via the AMPK pathway. eEF2K plays important roles in numerous biological systems, including neurology, cardiology, myology, and immunology. This review provides further insights into the current roles of eEF2K and its potential to be explored as a therapeutic target for drug development.
Collapse
Affiliation(s)
- Darby J. Ballard
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Hao-Yun Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Yijie Ren
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
12
|
Terrey M, Adamson SI, Chuang JH, Ackerman SL. Defects in translation-dependent quality control pathways lead to convergent molecular and neurodevelopmental pathology. eLife 2021; 10:e66904. [PMID: 33899734 PMCID: PMC8075583 DOI: 10.7554/elife.66904] [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: 01/26/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
Translation-dependent quality control pathways such as no-go decay (NGD), non-stop decay (NSD), and nonsense-mediated decay (NMD) govern protein synthesis and proteostasis by resolving non-translating ribosomes and preventing the production of potentially toxic peptides derived from faulty and aberrant mRNAs. However, how translation is altered and the in vivo defects that arise in the absence of these pathways are poorly understood. Here, we show that the NGD/NSD factors Pelo and Hbs1l are critical in mice for cerebellar neurogenesis but expendable for survival of these neurons after development. Analysis of mutant mouse embryonic fibroblasts revealed translational pauses, alteration of signaling pathways, and translational reprogramming. Similar effects on signaling pathways, including mTOR activation, the translatome and mouse cerebellar development were observed upon deletion of the NMD factor Upf2. Our data reveal that these quality control pathways that function to mitigate errors at distinct steps in translation can evoke similar cellular responses.
Collapse
Affiliation(s)
- Markus Terrey
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, Division of Biological Sciences, University of California San DiegoLa JollaUnited States
- Graduate School of Biomedical Sciences and Engineering, University of MaineOronoUnited States
| | - Scott I Adamson
- The Jackson Laboratory for Genomic MedicineFarmingtonUnited States
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn HealthFarmingtonUnited States
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic MedicineFarmingtonUnited States
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn HealthFarmingtonUnited States
| | - Susan L Ackerman
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, Division of Biological Sciences, University of California San DiegoLa JollaUnited States
| |
Collapse
|
13
|
Yang YP, Qin RH, Zhao JJ, Qin XY. BOP1 Silencing Suppresses Gastric Cancer Proliferation through p53 Modulation. Curr Med Sci 2021; 41:287-296. [PMID: 33877544 DOI: 10.1007/s11596-021-2345-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/26/2020] [Indexed: 10/21/2022]
Abstract
Block of proliferation 1 (BOP1) is a key protein involved in ribosome maturation and affects cancer progression. However, its role in gastric cancer (GC) remains unknown. This study aimed to explore the expression of BOP1 in GC and its potential mechanisms in regulating GC growth, and the relationship between BOP1 level in cancer tissues and survival was also analyzed. The expression of BOP1 was examined by immunohistochemistry (IHC) in a cohort containing 387 patients with primary GC. Cultured GC cells were treated by siRNA to knock down the BOP1 expression, and examined by CCK-8 assay and plate clone formation to assess cell proliferation in vitro. Apoptotic rate of cultured GC cells was detected by flow cytometry with double staining of AnnxinV/PI. The xenografted mouse model was used to assess GC cell proliferation in vivo. Western blot and IHC were also performed to detect the expression levels of BOP1, p53 and p21. Patients with higher level of BOP1 in cancer tissues had significantly poorer survival. BOP1 silencing significantly suppressed GC cell proliferation both in vitro and in vivo. It blocked cell cycle at G0/G1 phase and led to apoptosis of GC cells via upregulating p53 and p21. BOP1 silencing-induced suppression of cell proliferation was partly reversed by pifithrin-α (a p53 inhibitor). Our study demonstrated that BOP1 up-regulation may be a hallmark of GC and it may regulate proliferation of GC cells by activating p53. BOP1 might be considered a novel biomarker of GC proliferation, and could be a potential indicator of prognosis of GC patients. BOP1 might also be a potential target for the treatment of GC patients if further researched.
Collapse
Affiliation(s)
- Yu-Peng Yang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rui-Huan Qin
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Jun-Jie Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin-Yu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
BOP1 Knockdown Attenuates Neointimal Hyperplasia by Activating p53 and Inhibiting Nascent Protein Synthesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5986260. [PMID: 33510838 PMCID: PMC7826231 DOI: 10.1155/2021/5986260] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/18/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
The rate of ribosome biogenesis plays a vital role in cell cycle progression and proliferation and is strongly connected with coronary restenosis and atherosclerosis. Blocking of proliferation 1 (BOP1) has been found as an evolutionarily conserved gene and a pivotal regulator of ribosome biogenesis and cell proliferation. However, little is known about its role in neointimal formation and its relationship with vascular smooth muscle cell (VSMC) proliferation and migration. The present study mainly explores the effect of BOP1 on VSMCs, the progression of neointimal hyperplasia, and the pathogenic mechanism. The expression of BOP1 was found to be significantly elevated during neointimal formation in human coronary samples and the rat balloon injury model. BOP1 knockdown inspires the nucleolus stress, which subsequently activates the p53-dependent stress response pathway, and inhibits the nascent protein synthesis, which subsequently inhibits the proliferation and migration of VSMCs. Knockdown ribosomal protein L11 (RPL11) by transfecting with siRNA or inhibiting p53 by pifithrin-α (PFT-α) partly reserved the biological effects induced by BOP1 knockdown. The present study revealed that BOP1 deletion attenuates VSMC proliferation and migration by activating the p53-dependent nucleolus stress response pathway and inhibits the synthesis of nascent proteins. BOP1 may become a novel biological target for neointimal hyperplasia.
Collapse
|
15
|
Campos RK, Wijeratne HRS, Shah P, Garcia-Blanco MA, Bradrick SS. Ribosomal stalk proteins RPLP1 and RPLP2 promote biogenesis of flaviviral and cellular multi-pass transmembrane proteins. Nucleic Acids Res 2020; 48:9872-9885. [PMID: 32890404 PMCID: PMC7515724 DOI: 10.1093/nar/gkaa717] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/05/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
The ribosomal stalk proteins, RPLP1 and RPLP2 (RPLP1/2), which form the ancient ribosomal stalk, were discovered decades ago but their functions remain mysterious. We had previously shown that RPLP1/2 are exquisitely required for replication of dengue virus (DENV) and other mosquito-borne flaviviruses. Here, we show that RPLP1/2 function to relieve ribosome pausing within the DENV envelope coding sequence, leading to enhanced protein stability. We evaluated viral and cellular translation in RPLP1/2-depleted cells using ribosome profiling and found that ribosomes pause in the sequence coding for the N-terminus of the envelope protein, immediately downstream of sequences encoding two adjacent transmembrane domains (TMDs). We also find that RPLP1/2 depletion impacts a ribosome density for a small subset of cellular mRNAs. Importantly, the polarity of ribosomes on mRNAs encoding multiple TMDs was disproportionately affected by RPLP1/2 knockdown, implying a role for RPLP1/2 in multi-pass transmembrane protein biogenesis. These analyses of viral and host RNAs converge to implicate RPLP1/2 as functionally important for ribosomes to elongate through ORFs encoding multiple TMDs. We suggest that the effect of RPLP1/2 at TMD associated pauses is mediated by improving the efficiency of co-translational folding and subsequent protein stability.
Collapse
Affiliation(s)
- Rafael K Campos
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Premal Shah
- Department of Genetics, Rutgers University, NJ, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.,Programme of Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
16
|
MAPK-interacting kinase 2 (MNK2) regulates adipocyte metabolism independently of its catalytic activity. Biochem J 2020; 477:2735-2754. [DOI: 10.1042/bcj20200433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 11/17/2022]
Abstract
The mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs) are serine/threonine protein kinases that are activated by the ERK1/2 (extracellular regulated kinase) and p38α/β MAPK pathways. The MNKs have previously been implicated in metabolic disease and shown to mediate diet-induced obesity. In particular, knockout of MNK2 in mice protects from the weight gain induced by a high-fat diet. These and other data suggest that MNK2 regulates the expansion of adipose tissue (AT), a stable, long-term energy reserve that plays an important role in regulating whole-body energy homeostasis. Using the well-established mouse 3T3-L1 in vitro model of adipogenesis, the role of the MNKs in adipocyte differentiation and lipid storage was investigated. Inhibition of MNK activity using specific inhibitors failed to impair adipogenesis or lipid accumulation, suggesting that MNK activity is not required for adipocyte differentiation and does not regulate lipid storage. However, small-interfering RNA (siRNA) knock-down of MNK2 did reduce lipid accumulation and regulated the levels of two major lipogenic transcriptional regulators, ChREBP (carbohydrate response element-binding protein) and LPIN1 (Lipin-1). These factors are responsible for controlling the expression of genes for proteins involved in de novo lipogenesis and triglyceride synthesis. The knock-down of MNK2 also increased the expression of hormone-sensitive lipase which catalyses the breakdown of triglyceride. These findings identify MNK2 as a regulator of adipocyte metabolism, independently of its catalytic activity, and reveal some of the mechanisms by which MNK2 drives AT expansion. The development of an MNK2-targeted therapy may, therefore, be a useful intervention for reducing weight caused by excessive nutrient intake.
Collapse
|
17
|
Sandeman LY, Kang WX, Wang X, Jensen KB, Wong D, Bo T, Gao L, Zhao J, Byrne CD, Page AJ, Proud CG. Disabling MNK protein kinases promotes oxidative metabolism and protects against diet-induced obesity. Mol Metab 2020; 42:101054. [PMID: 32712434 PMCID: PMC7476876 DOI: 10.1016/j.molmet.2020.101054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives Diet-driven obesity is increasingly widespread. Its consequences pose major challenges to human health and health care systems. There are MAP kinase-interacting kinases (MNKs) in mice, MNK1 and MNK2. Studies have demonstrated that mice lacking either MNK1 or MNK2 were partially protected against high-fat diet (HFD)-induced weight gain and insulin resistance. The aims of this study were to evaluate the phenotype of mice lacking both MNKs when given an HFD, to assess whether pharmacological inhibition of MNK function also protects against diet-induced obesity (DIO) and its consequences and to probe the mechanisms underlying such protection. Methods Male wild-type (WT) C57Bl6 mice or mice lacking both MNK1 and MNK2 (double knockout, DKO) were fed an HFD or control diet (CD) for up to 16 weeks. In a separate study, WT mice were also given an HFD for 6 weeks, after which half were treated with the recently-developed MNK inhibitor ETC-206 daily for 10 more weeks while continuing an HFD. Metabolites and other parameters were measured, and the expression of selected mRNAs and proteins was assessed. Results MNK-DKO mice were almost completely protected from HFD-induced obesity. Higher energy expenditure (EE) in MNK-DKO mice was observed, which probably reflects the changes in a number of genes or proteins linked to lipolysis, mitochondrial function/biogenesis, oxidative metabolism, and/or ATP consumption. The MNK inhibitor ETC-206 also prevented HFD-induced weight gain, confirming that the activity of the MNKs facilitates weight gain due to excessive caloric consumption. Conclusions Disabling MNKs in mice, either genetically or pharmacologically, strongly prevents weight gain on a calorie-rich diet. This finding likely results from increased energy utilisation, involving greater ATP consumption, mitochondrial oxidative metabolism, and other processes. Knockout of MNK1/MNK2 protects mice against diet-induced obesity. MNK1/2 DKO mice have higher energy expenditure. MNK1/2 DKO increases the expression of genes of lipid and mitochondrial metabolism. Pharmacological inhibition of MNKs has similar effects.
Collapse
Affiliation(s)
- Lauren Y Sandeman
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Wan Xian Kang
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Kirk B Jensen
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Derick Wong
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Tao Bo
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia; Shandong-South Australia Joint Laboratory of Metabolic Disease Research, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ling Gao
- Shandong-South Australia Joint Laboratory of Metabolic Disease Research, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Shandong-South Australia Joint Laboratory of Metabolic Disease Research, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Christopher D Byrne
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, Hampshire, SO16 6YD, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, Hampshire, SO17 1BJ, UK
| | - Amanda J Page
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia; Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Diseases, Adelaide Medical School, Adelaide, SA, 5000, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
18
|
Okur MN, Lee JH, Osmani W, Kimura R, Demarest TG, Croteau DL, Bohr VA. Cockayne syndrome group A and B proteins function in rRNA transcription through nucleolin regulation. Nucleic Acids Res 2020; 48:2473-2485. [PMID: 31970402 PMCID: PMC7049711 DOI: 10.1093/nar/gkz1242] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023] Open
Abstract
Cockayne Syndrome (CS) is a rare neurodegenerative disease characterized by short stature, accelerated aging and short lifespan. Mutations in two human genes, ERCC8/CSA and ERCC6/CSB, are causative for CS and their protein products, CSA and CSB, while structurally unrelated, play roles in DNA repair and other aspects of DNA metabolism in human cells. Many clinical and molecular features of CS remain poorly understood, and it was observed that CSA and CSB regulate transcription of ribosomal DNA (rDNA) genes and ribosome biogenesis. Here, we investigate the dysregulation of rRNA synthesis in CS. We report that Nucleolin (Ncl), a nucleolar protein that regulates rRNA synthesis and ribosome biogenesis, interacts with CSA and CSB. In addition, CSA induces ubiquitination of Ncl, enhances binding of CSB to Ncl, and CSA and CSB both stimulate the binding of Ncl to rDNA and subsequent rRNA synthesis. CSB and CSA also increase RNA Polymerase I loading to the coding region of the rDNA and this is Ncl dependent. These findings suggest that CSA and CSB are positive regulators of rRNA synthesis via Ncl regulation. Most CS patients carry mutations in CSA and CSB and present with similar clinical features, thus our findings provide novel insights into disease mechanism.
Collapse
Affiliation(s)
- Mustafa N Okur
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jong-Hyuk Lee
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Wasif Osmani
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Risako Kimura
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Tyler G Demarest
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Danish Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
19
|
Yoshikawa M, Morifuji T, Matsumoto T, Maeshige N, Tanaka M, Fujino H. Effects of combined treatment with blood flow restriction and low-current electrical stimulation on muscle hypertrophy in rats. J Appl Physiol (1985) 2019; 127:1288-1296. [PMID: 31556832 DOI: 10.1152/japplphysiol.00070.2019] [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] [Indexed: 11/22/2022] Open
Abstract
This study aimed to clarify the effects of a combined treatment comprising blood flow restriction and low-current electrical stimulation on skeletal muscle hypertrophy in rats. Male Wistar rats were divided into control (Cont), blood flow restriction (Bfr), electrical stimulation (Es), or Bfr with Es (Bfr + Es) groups. Pressure cuffs (80 mmHg) were placed around the thighs of Bfr and Bfr + Es rats. Low-current Es was applied to calf muscles in the Es and Bfr + Es rats. In experiment 1, a 1-day treatment regimen (5-min stimulation, followed by 5-min rest) was delivered four times to study the acute effects. In experiment 2, the same treatment regimen was delivered three times/wk for 8 wk. Body weight, muscle mass, changes in maximal isometric contraction, fiber cross-sectional area of the soleus muscle, expression of phosphorylated and total-ERK1/2, phosphorylated-rpS6 Ser235/236, phosphorylated and total Akt, and phosphorylated-rpS6 Ser240/244 were measured. Bfr and Es treatment alone failed to induce muscle hypertrophy and increase the expression of phosphorylated rpS6 Ser240/244. Combined Bfr + Es upregulated muscle mass, increased the fiber cross-sectional area, and increased phosphorylated rpS6 Ser240/244 expression and phosphorylated rpS6 Ser235/236 expression compared with controls. Combined treatment with Bfr and low-current Es can induce muscle hypertrophy via activation of two protein synthesis signaling pathways. This treatment should be introduced for older patients with sarcopenia and others with muscle weakness.NEW & NOTEWORTHY We investigated the acute and chronic effect of low-current electrical stimulation with blood flow restriction on skeletal muscle hypertrophy and the mechanisms controlling the hypertrophic response. Low-current electrical stimulation could not induce skeletal muscle hypertrophy, but a combination treatment did. Blood lactate and growth hormone levels were increased in the early response. Moreover, activation of ERK1/2 and mTOR pathways were observed in both the acute and chronic response, which contribute to muscle hypertrophy.
Collapse
Affiliation(s)
- Madoka Yoshikawa
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan.,Department of Rehabilitation Science, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Tomohiro Matsumoto
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| |
Collapse
|
20
|
Kearse MG, Goldman DH, Choi J, Nwaezeapu C, Liang D, Green KM, Goldstrohm AC, Todd PK, Green R, Wilusz JE. Ribosome queuing enables non-AUG translation to be resistant to multiple protein synthesis inhibitors. Genes Dev 2019; 33:871-885. [PMID: 31171704 PMCID: PMC6601509 DOI: 10.1101/gad.324715.119] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/24/2019] [Indexed: 02/05/2023]
Abstract
Aberrant translation initiation at non-AUG start codons is associated with multiple cancers and neurodegenerative diseases. Nevertheless, how non-AUG translation may be regulated differently from canonical translation is poorly understood. Here, we used start codon-specific reporters and ribosome profiling to characterize how translation from non-AUG start codons responds to protein synthesis inhibitors in human cells. These analyses surprisingly revealed that translation of multiple non-AUG-encoded reporters and the endogenous GUG-encoded DAP5 (eIF4G2/p97) mRNA is resistant to cycloheximide (CHX), a translation inhibitor that severely slows but does not completely abrogate elongation. Our data suggest that slowly elongating ribosomes can lead to queuing/stacking of scanning preinitiation complexes (PICs), preferentially enhancing recognition of weak non-AUG start codons. Consistent with this model, limiting PIC formation or scanning sensitizes non-AUG translation to CHX. We further found that non-AUG translation is resistant to other inhibitors that target ribosomes within the coding sequence but not those targeting newly initiated ribosomes. Together, these data indicate that ribosome queuing enables mRNAs with poor initiation context-namely, those with non-AUG start codons-to be resistant to pharmacological translation inhibitors at concentrations that robustly inhibit global translation.
Collapse
Affiliation(s)
- Michael G Kearse
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Daniel H Goldman
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jiou Choi
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Chike Nwaezeapu
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Dongming Liang
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Katelyn M Green
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Aaron C Goldstrohm
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Veterans Affairs Medical Center, Ann Arbor, Michigan 48105, USA
| | - Rachel Green
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jeremy E Wilusz
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
21
|
von Walden F. Ribosome biogenesis in skeletal muscle: coordination of transcription and translation. J Appl Physiol (1985) 2019; 127:591-598. [PMID: 31219775 DOI: 10.1152/japplphysiol.00963.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle mass responds in a remarkable manner to alterations in loading and use. It has long been clear that skeletal muscle hypertrophy can be prevented by inhibiting RNA synthesis. Since 80% of the cell's total RNA has been estimated to be rRNA, this finding indicates that de novo production of rRNA via transcription of the corresponding genes is important for such hypertrophy to occur. Transcription of rDNA by RNA Pol I is the rate-limiting step in ribosome biogenesis, indicating in turn that this biogenesis strongly influences the hypertrophic response. The present minireview focuses on 1) a brief description of the key steps in ribosome biogenesis and the relationship of this process to skeletal muscle mass and 2) the coordination of ribosome biogenesis and protein synthesis for growth or atrophy, as exemplified by the intracellular AMPK and mTOR pathways.
Collapse
Affiliation(s)
- Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
22
|
Abnormal Ribosome Biogenesis Partly Induced p53-Dependent Aortic Medial Smooth Muscle Cell Apoptosis and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7064319. [PMID: 31210846 PMCID: PMC6532287 DOI: 10.1155/2019/7064319] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/30/2019] [Accepted: 04/14/2019] [Indexed: 12/19/2022]
Abstract
Ribosome biogenesis is a crucial biological process related to cell proliferation, redox balance, and muscle contractility. Aortic smooth muscle cells (ASMCs) show inhibition of proliferation and apoptosis, along with high levels of oxidative stress in aortic dissection (AD). Theoretically, ribosome biogenesis should be enhanced in the ASMCs at its proliferative state but suppressed during apoptosis and oxidative stress. However, the exact status and role of ribosome biogenesis in AD are unknown. We therefore analyzed the expression levels of BOP1, a component of the PeBoW complex which is crucial to ribosome biogenesis, in AD patients and a murine AD model and its influence on the ASMCs. BOP1 was downregulated in the aortic tissues of AD patients compared to healthy donors. In addition, overexpression of BOP1 in human aortic smooth muscle cells (HASMCs) inhibited apoptosis and accumulation of p53 under hypoxic conditions, while knockdown of BOP1 decreased the protein synthesis rate and motility of HASMCs. The RNA polymerase I inhibitor cx-5461 induced apoptosis, ROS production, and proliferative inhibition in the HASMCs, which was partly attenuated by p53 knockout. Furthermore, cx-5461 aggravated the severity of AD in vivo, but a p53-/- background extended the life-span and lowered AD incidence in the mice. Taken together, decreased ribosome biogenesis in ASMCs resulting in p53-dependent proliferative inhibition, oxidative stress, and apoptosis is one of the underlying mechanisms of AD.
Collapse
|
23
|
Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11:326-334. [PMID: 30225988 DOI: 10.1111/1753-0407.12857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) results in decreased muscle mass and harmful complications. Blood flow restriction (Bfr) and electrical stimulation (ES) increase muscle protein synthesis. We hypothesized that combined Bfr and low-intensity ES may be more effective in preventing diabetes-associated muscle atrophy by inhibiting the downregulation of protein synthesis-related pathways. In this study, the effects of combined Bfr and low-intensity ES on diabetes-associated muscle atrophy were investigated by evaluating advanced glycation end-products (AGEs) and receptor for AGEs (RAGE) in rats. METHODS Twenty-four Goto-Kakizaki (GK) rats were randomly divided into four treatment groups: sedentary DM, DM + Bfr (pressure cuffs placed around the thigh), DM + ES, and DM + Bfr + ES. Six Wistar rats were used as an age-matched control. Levels of AGEs and the expression of RAGE, extracellular signal-regulated kinase (ERK), and ribosomal protein S6 (rpS6) were determined in plantaris muscles. RESULTS Diabetes resulted in a loss of muscle mass and upregulation of AGEs and RAGE in the plantaris muscle compared with the control group. Treatment with Bfr and ES alone failed to attenuate diabetes-associated loss of muscle mass, and could not prevent the upregulation of AGEs. However, the combination of Bfr and ES prevented the diabetes-associated decrease in muscle mass and upregulation of AGEs. In addition, the combination treatment prevented diabetes-associated decreases in the expression of phosphorylated rpS6. CONCLUSIONS Combination treatment with Bfr and ES may prevent diabetes-associated muscle atrophy by upregulating inhibition of AGEs, which leads to the activation of protein synthesis.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Madoka Yoshikawa
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation, Kobe International University, Kobe, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
24
|
Bakshi A, Moin M, Madhav MS, Kirti PB. Target of rapamycin, a master regulator of multiple signalling pathways and a potential candidate gene for crop improvement. PLANT BIOLOGY (STUTTGART, GERMANY) 2019; 21:190-205. [PMID: 30411830 DOI: 10.1111/plb.12935] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/05/2018] [Indexed: 06/08/2023]
Abstract
The target of rapamycin (TOR) protein regulates growth and development in photosynthetic and non-photosynthetic eukaryotes. Although the TOR regulatory networks are involved in nutrient and energy signalling, and transcriptional and translational control of multiple signalling pathways, the molecular mechanism of TOR regulation of plant abiotic stress responses is still unclear. The TOR-mediated transcriptional regulation of genes encoding ribosomal proteins (RP) is a necessity under stress conditions for balanced growth and productivity in plants. The activation of SnRKs (sucrose non-fermenting-related kinases) and the inactivation of TOR signalling in abiotic stresses is in line with the accumulation of ABA and transcriptional activation of stress responsive genes. Autophagy is induced under abiotic stress conditions, which results in degradation of proteins and the release of amino acids, which might possibly induce phosphorylation of TOR and, hence, its activation. TOR signalling also has a role in regulating ABA biosynthesis for transcriptional regulation of stress-related genes. The switch between activation and inactivation of TOR by its phosphorylation and de-phosphorylation maintains balanced growth in response to stresses. In the present review, we discuss the important signalling pathways that are regulated by TOR and try to assess the relationship between TOR signalling and tolerance to abiotic stresses in plants. The review also discusses possible cross-talk between TOR and RP genes in response to abiotic stresses.
Collapse
Affiliation(s)
- A Bakshi
- Department of Plant Sciences, University of Hyderabad, Hyderabad, India
| | - M Moin
- Department of Biotechnology, Indian Institute of Rice Research, Hyderabad, India
| | - M S Madhav
- Department of Biotechnology, Indian Institute of Rice Research, Hyderabad, India
| | - P B Kirti
- Department of Plant Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
25
|
Mirzoev TM, Shenkman BS. Regulation of Protein Synthesis in Inactivated Skeletal Muscle: Signal Inputs, Protein Kinase Cascades, and Ribosome Biogenesis. BIOCHEMISTRY (MOSCOW) 2018; 83:1299-1317. [PMID: 30482143 DOI: 10.1134/s0006297918110020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Disuse atrophy of skeletal muscles is characterized by a significant decrease in the mass and size of muscle fibers. Disuse atrophy develops as a result of prolonged reduction in the muscle functional activity caused by bed rest, limb immobilization, and real or simulated microgravity. Disuse atrophy is associated with the downregulation of protein biosynthesis and simultaneous activation of protein degradation. This review is focused on the key molecular mechanisms regulating the rate of protein synthesis in mammalian skeletal muscles during functional unloading.
Collapse
Affiliation(s)
- T M Mirzoev
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia.
| | - B S Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia
| |
Collapse
|
26
|
Rotblat B, Agostini M, Niklison-Chirou MV, Amelio I, Willis AE, Melino G. Sustained protein synthesis and reduced eEF2K levels in TAp73 -\- mice brain: a possible compensatory mechanism. Cell Cycle 2018; 17:2637-2643. [PMID: 30507330 DOI: 10.1080/15384101.2018.1553341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The transcription factor p73 is a member of the p53 family, of which the transactivation domain containing isoform (TAp73) plays key roles in brain development and neuronal stem cells. TAp73 also facilitates homoeostasis and prevents oxidative damage in vivo by inducing the expression of its target genes. Recently, we found that in addition to its role in regulation of transcription, TAp73 also affects mRNA translation. In cultured cells, acute TAp73 depletion activates eEF2K, which phosphorylates eEF2 reducing mRNA translation elongation. As a consequence, there is a reduction in global proteins synthesis rates and reprogramming of the translatome, leading to a selective decrease in the translation of rRNA processing factors. Given the dramatic effects of Tap73 depletion in vitro it was important to determine whether similar effects were observed in vivo. Here, we report the surprising finding that in brains of TAp73 KO mice there is a reduced level of eEF2K, which allows protein synthesis rates to be maintained suggesting a compensation model. These data provide new insights to the role of TAp73 in translation regulation and the eEF2K pathway in the brain.
Collapse
Affiliation(s)
- Barak Rotblat
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,b Department of Life Sciences , Ben Gurion University in the Negev , Beer Sheva , Israel
| | - Massimiliano Agostini
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| | - Maria Victoria Niklison-Chirou
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,d Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry , Queen Mary University of London , London , UK
| | - Ivano Amelio
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Anne E Willis
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Gerry Melino
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| |
Collapse
|
27
|
Marini A, Rotblat B, Sbarrato T, Niklison-Chirou MV, Knight JRP, Dudek K, Jones C, Bushell M, Knight RA, Amelio I, Willis AE, Melino G. TAp73 contributes to the oxidative stress response by regulating protein synthesis. Proc Natl Acad Sci U S A 2018; 115:6219-6224. [PMID: 29844156 PMCID: PMC6004440 DOI: 10.1073/pnas.1718531115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TAp73 is a transcription factor that plays key roles in brain development, aging, and cancer. At the cellular level, TAp73 is a critical homeostasis-maintaining factor, particularly following oxidative stress. Although major studies focused on TAp73 transcriptional activities have indicated a contribution of TAp73 to cellular metabolism, the mechanisms underlying its role in redox homeostasis have not been completely elucidated. Here we show that TAp73 contributes to the oxidative stress response by participating in the control of protein synthesis. Regulation of mRNA translation occupies a central position in cellular homeostasis during the stress response, often by reducing global rates of protein synthesis and promoting translation of specific mRNAs. TAp73 depletion results in aberrant ribosomal RNA (rRNA) processing and impaired protein synthesis. In particular, polysomal profiles show that TAp73 promotes the integration of mRNAs that encode rRNA-processing factors in polysomes, supporting their translation. Concurrently, TAp73 depletion causes increased sensitivity to oxidative stress that correlates with reduced ATP levels, hyperactivation of AMPK, and translational defects. TAp73 is important for maintaining active translation of mitochondrial transcripts in response to oxidative stress, thus promoting mitochondrial activity. Our results indicate that TAp73 contributes to redox homeostasis by affecting the translational machinery, facilitating the translation of specific mitochondrial transcripts. This study identifies a mechanism by which TAp73 contributes to the oxidative stress response and describes a completely unexpected role for TAp73 in regulating protein synthesis.
Collapse
Affiliation(s)
- Alberto Marini
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Barak Rotblat
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Thomas Sbarrato
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Maria Victoria Niklison-Chirou
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - John R P Knight
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Kate Dudek
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Carolyn Jones
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Martin Bushell
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Richard A Knight
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom
| | - Anne E Willis
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom;
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, United Kingdom;
- Department of Experimental Medicine and Surgery, IDI-IRCCS (Istituto Dermopatico dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico), University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
28
|
Liakath-Ali K, Mills EW, Sequeira I, Lichtenberger BM, Pisco AO, Sipilä KH, Mishra A, Yoshikawa H, Wu CCC, Ly T, Lamond AI, Adham IM, Green R, Watt FM. An evolutionarily conserved ribosome-rescue pathway maintains epidermal homeostasis. Nature 2018; 556:376-380. [PMID: 29643507 DOI: 10.1038/s41586-018-0032-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 02/28/2018] [Indexed: 01/01/2023]
Abstract
Ribosome-associated mRNA quality control mechanisms ensure the fidelity of protein translation1,2. Although these mechanisms have been extensively studied in yeast, little is known about their role in mammalian tissues, despite emerging evidence that stem cell fate is controlled by translational mechanisms3,4. One evolutionarily conserved component of the quality control machinery, Dom34 (in higher eukaryotes known as Pelota (Pelo)), rescues stalled ribosomes 5 . Here we show that Pelo is required for mammalian epidermal homeostasis. Conditional deletion of Pelo in mouse epidermal stem cells that express Lrig1 results in hyperproliferation and abnormal differentiation of these cells. By contrast, deletion of Pelo in Lgr5-expressing stem cells has no effect and deletion in Lgr6-expressing stem cells induces only a mild phenotype. Loss of Pelo results in accumulation of short ribosome footprints and global upregulation of translation, rather than affecting the expression of specific genes. Translational inhibition by rapamycin-mediated downregulation of mTOR (mechanistic target of rapamycin kinase) rescues the epidermal phenotype. Our study reveals that the ribosome-rescue machinery is important for mammalian tissue homeostasis and that it has specific effects on different stem cell populations.
Collapse
Affiliation(s)
- Kifayathullah Liakath-Ali
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA, USA
| | - Eric W Mills
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Inês Sequeira
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Beate M Lichtenberger
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Kalle H Sipilä
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Ajay Mishra
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
- Cambridge Infinitus Research Centre, University of Cambridge, Cambridge, UK
| | - Harunori Yoshikawa
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Colin Chih-Chien Wu
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tony Ly
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Ibrahim M Adham
- Institute of Human Genetics, University Medical Centre of Göttingen, Göttingen, Germany
| | - Rachel Green
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK.
| |
Collapse
|
29
|
El-Naggar AM, Sorensen PH. Translational control of aberrant stress responses as a hallmark of cancer. J Pathol 2018; 244:650-666. [PMID: 29293271 DOI: 10.1002/path.5030] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/12/2022]
Abstract
Altered mRNA translational control is emerging as a critical factor in cancer development and progression. Targeting specific elements of the translational machinery, such as mTORC1 or eIF4E, is emerging as a new strategy for innovative cancer therapy. While translation of most mRNAs takes place through cap-dependent mechanisms, a sub-population of cellular mRNA species, particularly stress-inducible mRNAs with highly structured 5'-UTR regions, are primarily translated through cap-independent mechanisms. Intriguingly, many of these mRNAs encode proteins that are involved in tumour cell adaptation to microenvironmental stress, and thus linked to aggressive behaviour including tumour invasion and metastasis. This necessitates a rigorous search for links between microenvironmental stress and aggressive tumour phenotypes. Under stress, cells block global protein synthesis to preserve energy while maintaining selective synthesis of proteins that support cell survival. One highly conserved mechanism to regulate protein synthesis under cell stress is to sequester mRNAs into cytosolic aggregates called stress granules (SGs), where their translation is silenced. SGs confer survival advantages and chemotherapeutic resistance to tumour cells under stress. Recently, it has been shown that genetically blocking SG formation dramatically reduces tumour invasive and metastatic capacity in vivo. Therefore, targeting SG formation might represent a potential treatment strategy to block cancer metastasis. Here, we present the critical link between selective mRNA translation, stress adaptation, SGs, and tumour progression. Further, we also explain how deciphering mechanisms of selective mRNA translation occurs under cell stress holds great promise for the identification of new targets in the treatment of cancer. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Amal M El-Naggar
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, Canada.,Department of Pathology, Faculty of Medicine, Menoufia University, Egypt
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, Canada
| |
Collapse
|
30
|
Sapio RT, Nezdyur AN, Krevetski M, Anikin L, Manna VJ, Minkovsky N, Pestov DG. Inhibition of post-transcriptional steps in ribosome biogenesis confers cytoprotection against chemotherapeutic agents in a p53-dependent manner. Sci Rep 2017; 7:9041. [PMID: 28831158 PMCID: PMC5567254 DOI: 10.1038/s41598-017-09002-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022] Open
Abstract
The p53-mediated nucleolar stress response associated with inhibition of ribosomal RNA transcription was previously shown to potentiate killing of tumor cells. Here, we asked whether targeting of ribosome biogenesis can be used as the basis for selective p53-dependent cytoprotection of nonmalignant cells. Temporary functional inactivation of the 60S ribosome assembly factor Bop1 in a 3T3 cell model markedly increased cell recovery after exposure to camptothecin or methotrexate. This was due, at least in part, to reversible pausing of the cell cycle preventing S phase associated DNA damage. Similar cytoprotective effects were observed after transient shRNA-mediated silencing of Rps19, but not several other tested ribosomal proteins, indicating distinct cellular responses to the inhibition of different steps in ribosome biogenesis. By temporarily inactivating Bop1 function, we further demonstrate selective killing of p53-deficient cells with camptothecin while sparing isogenic p53-positive cells. Thus, combining cytotoxic treatments with inhibition of select post-transcriptional steps of ribosome biogenesis holds potential for therapeutic targeting of cells that have lost p53.
Collapse
Affiliation(s)
- Russell T Sapio
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Anastasiya N Nezdyur
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, 08028, USA
| | - Matthew Krevetski
- Department of Biological Sciences, Rowan University, Glassboro, NJ, 08028, USA
| | - Leonid Anikin
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Vincent J Manna
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Natalie Minkovsky
- Department of Biological Sciences, Rowan University, Glassboro, NJ, 08028, USA
| | - Dimitri G Pestov
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.
| |
Collapse
|
31
|
Khattar E, Kumar P, Liu CY, Akıncılar SC, Raju A, Lakshmanan M, Maury JJP, Qiang Y, Li S, Tan EY, Hui KM, Shi M, Loh YH, Tergaonkar V. Telomerase reverse transcriptase promotes cancer cell proliferation by augmenting tRNA expression. J Clin Invest 2016; 126:4045-4060. [PMID: 27643433 DOI: 10.1172/jci86042] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 08/11/2016] [Indexed: 12/13/2022] Open
Abstract
Transcriptional reactivation of telomerase reverse transcriptase (TERT) reconstitutes telomerase activity in the majority of human cancers. Here, we found that ectopic TERT expression increases cell proliferation, while acute reductions in TERT levels lead to a dramatic loss of proliferation without any change in telomere length, suggesting that the effects of TERT could be telomere independent. We observed that TERT determines the growth rate of cancer cells by directly regulating global protein synthesis independently of its catalytic activity. Genome-wide TERT binding across 5 cancer cell lines and 2 embryonic stem cell lines revealed that endogenous TERT, driven by mutant promoters or oncogenes, directly associates with the RNA polymerase III (pol III) subunit RPC32 and enhances its recruitment to chromatin, resulting in increased RNA pol III occupancy and tRNA expression in cancers. TERT-deficient mice displayed marked delays in polyomavirus middle T oncogene-induced (PyMT-induced) mammary tumorigenesis, increased survival, and reductions in tRNA levels. Ectopic expression of either RPC32 or TERT restored tRNA levels and proliferation defects in TERT-depleted cells. Finally, we determined that levels of TERT and tRNA correlated in breast and liver cancer samples. Together, these data suggest the existence of a unifying mechanism by which TERT enhances translation in cells to regulate cancer cell proliferation.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Neoplastic/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- Mammary Neoplasms, Experimental/enzymology
- Mammary Neoplasms, Experimental/pathology
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasm Transplantation
- Promoter Regions, Genetic
- Protein Binding
- Protein Biosynthesis
- Protein Interaction Domains and Motifs
- RNA Polymerase III/metabolism
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- Telomerase/physiology
Collapse
|
32
|
Liu R, Kenney JW, Manousopoulou A, Johnston HE, Kamei M, Woelk CH, Xie J, Schwarzer M, Garbis SD, Proud CG. Quantitative Non-canonical Amino Acid Tagging (QuaNCAT) Proteomics Identifies Distinct Patterns of Protein Synthesis Rapidly Induced by Hypertrophic Agents in Cardiomyocytes, Revealing New Aspects of Metabolic Remodeling. Mol Cell Proteomics 2016; 15:3170-3189. [PMID: 27512079 PMCID: PMC5054342 DOI: 10.1074/mcp.m115.054312] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 01/16/2023] Open
Abstract
Cardiomyocytes undergo growth and remodeling in response to specific pathological or physiological conditions. In the former, myocardial growth is a risk factor for cardiac failure and faster protein synthesis is a major factor driving cardiomyocyte growth. Our goal was to quantify the rapid effects of different pro-hypertrophic stimuli on the synthesis of specific proteins in ARVC and to determine whether such effects are caused by alterations on mRNA abundance or the translation of specific mRNAs. Cardiomyocytes have very low rates of protein synthesis, posing a challenging problem in terms of studying changes in the synthesis of specific proteins, which also applies to other nondividing primary cells. To study the rates of accumulation of specific proteins in these cells, we developed an optimized version of the Quantitative Noncanonical Amino acid Tagging LC/MS proteomic method to label and selectively enrich newly synthesized proteins in these primary cells while eliminating the suppressive effects of pre-existing and highly abundant nonisotope-tagged polypeptides. Our data revealed that a classical pathologic (phenylephrine; PE) and the recently identified insulin stimulus that also contributes to the development of pathological cardiac hypertrophy (insulin), both increased the synthesis of proteins involved in, e.g. glycolysis, the Krebs cycle and beta-oxidation, and sarcomeric components. However, insulin increased synthesis of many metabolic enzymes to a greater extent than PE. Using a novel validation method, we confirmed that synthesis of selected candidates is indeed up-regulated by PE and insulin. Synthesis of all proteins studied was up-regulated by signaling through mammalian target of rapamycin complex 1 without changes in their mRNA levels, showing the key importance of translational control in the rapid effects of hypertrophic stimuli. Expression of PKM2 was up-regulated in rat hearts following TAC. This isoform possesses specific regulatory properties, so this finding indicates it may be involved in metabolic remodeling and also serve as a novel candidate biomarker. Levels of translation factor eEF1 also increased during TAC, likely contributing to faster cell mass accumulation. Interestingly those two candidates were not up-regulated in pregnancy or exercise induced CH, indicating PKM2 and eEF1 were pathological CH specific markers. We anticipate that the methodologies described here will be valuable for other researchers studying protein synthesis in primary cells.
Collapse
Affiliation(s)
- Rui Liu
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Justin W Kenney
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Antigoni Manousopoulou
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Harvey E Johnston
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Makoto Kamei
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Christopher H Woelk
- ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jianling Xie
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Michael Schwarzer
- **Department of Cardiovascular Surgery, Jena University Hospital-Friedrich Schiller University of Jena, Erlanger Allee 101, 07747 Jena, Germany
| | - Spiros D Garbis
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK;
| | - Christopher G Proud
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA5005, Australia
| |
Collapse
|
33
|
Moore CEJ, Pickford J, Cagampang FR, Stead RL, Tian S, Zhao X, Tang X, Byrne CD, Proud CG. MNK1 and MNK2 mediate adverse effects of high-fat feeding in distinct ways. Sci Rep 2016; 6:23476. [PMID: 27087055 PMCID: PMC4834573 DOI: 10.1038/srep23476] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/07/2016] [Indexed: 02/06/2023] Open
Abstract
The MAP kinase-interacting kinases (MNK1 and MNK2) are non-essential enzymes which are activated by MAP kinases. They are implicated in controlling protein synthesis. Here we show that mice in which the expression of either MNK1 or MNK2 has been knocked out (KO) are protected against adverse effects of high-fat feeding, and in distinct ways. High-fat diet (HFD)-fed MNK2-KO show less weight gain than wild-type animals, and improved glucose tolerance, better insulin sensitivity and markedly diminished adipose tissue inflammation. This suggests MNK2 plays a role in adipogenesis and/or lipogenesis and in macrophage biology. MNK1-KO/HFD mice show better glucose tolerance and insulin sensitivity, but gain weight and show similar adipose inflammation to WT animals. These data suggest MNK1 participates in mediating HFD-induced insulin resistance. Our findings reveal distinct roles for the MNKs in a novel area of disease biology, metabolic dysfunction, and suggests they are potential new targets for managing metabolic disease.
Collapse
Affiliation(s)
- C E J Moore
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - J Pickford
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - F R Cagampang
- Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton SO16 6YD, United Kingdom
| | - R L Stead
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - S Tian
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - X Zhao
- Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - X Tang
- Department of Biochemistry &Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058 China
| | - C D Byrne
- Nutrition and Metabolism, University of Southampton, Faculty of Medicine, Southampton SO16 6YD, United Kingdom.,Southampton National Institute for Health Research, Biomedical Research Centre, University Hospital, Southampton, UK
| | - C G Proud
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
34
|
Denisenko O, Lucas ES, Sun C, Watkins AJ, Mar D, Bomsztyk K, Fleming TP. Regulation of ribosomal RNA expression across the lifespan is fine-tuned by maternal diet before implantation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:906-13. [PMID: 27060415 PMCID: PMC4914606 DOI: 10.1016/j.bbagrm.2016.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/18/2016] [Accepted: 04/04/2016] [Indexed: 01/17/2023]
Abstract
Cells and organisms respond to nutrient deprivation by decreasing global rates of transcription, translation and DNA replication. To what extent such changes can be reversed is largely unknown. We examined the effect of maternal dietary restriction on RNA synthesis in the offspring. Low protein diet fed either throughout gestation or for the preimplantation period alone reduced cellular RNA content across fetal somatic tissues during challenge and increased it beyond controls in fetal and adult tissues after challenge release. Changes in transcription of ribosomal RNA, the major component of cellular RNA, were responsible for this phenotype as evidenced by matching alterations in RNA polymerase I density and DNA methylation at ribosomal DNA loci. Cellular levels of the ribosomal transcription factor Rrn3 mirrored the rRNA expression pattern. In cell culture experiments, Rrn3 overexpression reduced rDNA methylation and increased rRNA expression; the converse occurred after inhibition of Rrn3 activity. These observations define novel mechanism where poor nutrition before implantation irreversibly alters basal rates of rRNA transcription thereafter in a process mediated by rDNA methylation and Rrn3 factor. Maternal malnutrition downregulates rDNA transcription in fetal tissues. Switch to normal diet permanently upregulates rDNA transcription compared to controls. These changes are mediated by DNA methylation and Pol I transcription factor Rrn3. This mechanism is activated before implantation.
Collapse
Affiliation(s)
- Oleg Denisenko
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA.
| | - Emma S Lucas
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Congshan Sun
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Adam J Watkins
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Daniel Mar
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA
| | - Karol Bomsztyk
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA
| | - Tom P Fleming
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
35
|
Wang X, Zhang Y, Jiang L, Zhou F, Zhai H, Zhang M, Wang J. Interpreting the distinct and shared genetic characteristics between Epstein-Barr virus associated and non-associated gastric carcinoma. Gene 2015; 576:798-806. [PMID: 26584536 DOI: 10.1016/j.gene.2015.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 12/26/2022]
Abstract
Gastric carcinoma is one of the major causes of cancer mortality worldwide. There is a better prognosis for patients with Epstein-Barr virus (EBV)-associated gastric carcinoma (EBVaGC) compared with those with EBV negative gastric carcinoma (EBVnGC). It is partly due to the fact that EBV infection recruits lymphocytes infiltrating the tumor. It has been reported that this infection indeed resulted in the changes in immune response genes and thus preventing the development of tumor. It is worthwhile to do a systematic study of EBVaGC and EBVnGC based on genetic characteristics and pathways. In this study, we investigated the information of gene ontology (GO) and KEGG pathway annotations to characterize EBVaGC and EBVnGC-related genes. By applying minimum redundancy maximum relevance (mRMR) algorithm, we provided an optimal set of features for identifying the EBVaGC and EBVnGC. We also employed the shortest path algorithm to probe the novel EBVaGC- and EBVnGC-related genes based on the interaction network of genes that differently expressed in them respectively. We obtained 1039 and 1003 features to identify these two types of gastric carcinoma respectively. Based on the optimal features of classification, we predicted 1881 and 2475 novel genes as additional candidates to support clinical research respectively for these two types of gastric cancers. We compared the differences and similarities of molecular traits between EBVaGC and EBVnGC, which would facilitate the understanding of gastric cancer and its therapy and was thus clinically relevant.
Collapse
Affiliation(s)
- Xixun Wang
- Department of Abodomenal Surgery, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Yifei Zhang
- Department of Abodomenal Surgery, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Lixin Jiang
- Department of Abodomenal Surgery, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Furun Zhou
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Huiyuan Zhai
- Department of Abodomenal Surgery, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Menglai Zhang
- Department of Abodomenal Surgery, Yantai Yuhuangding Hospital, Shandong, PR China
| | - Jinglin Wang
- Department of Emergency Center, Yantai Yuhuangding Hospital, Shandong, PR China.
| |
Collapse
|
36
|
Goudarzi KM, Nistér M, Lindström MS. mTOR inhibitors blunt the p53 response to nucleolar stress by regulating RPL11 and MDM2 levels. Cancer Biol Ther 2015; 15:1499-514. [PMID: 25482947 DOI: 10.4161/15384047.2014.955743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a master regulator of cell growth through its ability to stimulate ribosome biogenesis and mRNA translation. In contrast, the p53 tumor suppressor negatively controls cell growth and is activated by a wide range of insults to the cell. The mTOR and p53 signaling pathways are connected by a number of different mechanisms. Chemotherapeutics that inhibit ribosome biogenesis often induce nucleolar stress and activation of p53. Here we have investigated how the p53 response to nucleolar stress is affected by simultaneous mTOR inhibition in osteosarcoma and glioma cell lines. We found that inhibitors of the mTOR pathway including rapamycin, wortmannin, and caffeine blunted the p53 response to nucleolar stress induced by actinomycin D. Synthetic inhibitors of mTOR (temsirolimus, LY294.002 and PP242) also impaired actinomycin D triggered p53 stabilization and induction of p21. Ribosomal protein (RPL11) is known to be required for p53 protein stabilization following nucleolar stress. Treatment of cells with mTOR inhibitors may lead to reduced synthesis of RPL11 and thereby destabilize p53. We found that rapamycin mimicked the effect of RPL11 depletion in terms of blunting the p53 response to nucleolar stress. However, the extent to which the levels of p53 and RPL11 were reduced by rapamycin varied between cell lines. Additional mechanisms whereby rapamycin blunts the p53 response to nucleolar stress are likely to be involved. Indeed, rapamycin increased the levels of endogenous MDM2 despite inhibition of its phosphorylation at Ser-166. Our findings may have implications for the design of combinatorial cancer treatments with mTOR pathway inhibitors.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- Act D, actinomycin D
- BrdU, bromodeoxyuridine
- CHX, cycloheximide
- DMSO, dimethylsulphoxide
- DOX, doxorubicin
- EGCG, epigallocatechin-3-gallate
- FACS, fluorescence-activated cell sorting
- MPA, mycophenolic acid
- MTT, (3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromide)
- PI, propidium iodide
- actinomycin D
- caffeine
- glioma
- mTOR
- mTOR, mechanistic target of rapamycin
- nutlin-3
- p21
- p53
- rapamycin
- ribosomal protein L11
- ribosome biogenesis
Collapse
Affiliation(s)
- Kaveh M Goudarzi
- a Department of Oncology-Pathology; Karolinska Institutet; Cancer Center Karolinska ; Karolinska University Hospital ; Stockholm , Sweden
| | | | | |
Collapse
|
37
|
The DHX33 RNA Helicase Promotes mRNA Translation Initiation. Mol Cell Biol 2015; 35:2918-31. [PMID: 26100019 DOI: 10.1128/mcb.00315-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/15/2015] [Indexed: 12/29/2022] Open
Abstract
DEAD/DEAH box RNA helicases play essential roles in numerous RNA metabolic processes, such as mRNA translation, pre-mRNA splicing, ribosome biogenesis, and double-stranded RNA sensing. Herein we show that a recently characterized DEAD/DEAH box RNA helicase, DHX33, promotes mRNA translation initiation. We isolated intact DHX33 protein/RNA complexes in cells and identified several ribosomal proteins, translation factors, and mRNAs. Reduction of DHX33 protein levels markedly reduced polyribosome formation and caused the global inhibition of mRNA translation that was rescued with wild-type DHX33 but not helicase-defective DHX33. Moreover, we observed an accumulation of mRNA complexes with the 80S ribosome in the absence of functional DHX33, consistent with a stalling in initiation, and DHX33 more preferentially promoted structured mRNA translation. We conclude that DHX33 functions to promote elongation-competent 80S ribosome assembly at the late stage of mRNA translation initiation. Our results reveal a newly recognized function of DHX33 in mRNA translation initiation, further solidifying its central role in promoting cell growth and proliferation.
Collapse
|
38
|
The MAP kinase-interacting kinases regulate cell migration, vimentin expression and eIF4E/CYFIP1 binding. Biochem J 2015; 467:63-76. [DOI: 10.1042/bj20141066] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The MAP kinase-interacting kinases (Mnk1 and Mnk2) are activated by ERK and are best known for phosphorylating the translation initiation factor eIF4E. Genetic knockout of the Mnks impaired the migration of embryonic fibroblasts both in two-dimensional wound-healing experiments and in three-dimensional migration assays. Furthermore, a novel and selective Mnk inhibitor, Mnk-I1, which potently blocks eIF4E phosphorylation, blocked the migration of fibroblasts and cancer cells, without exerting ‘off-target’ effects on other signalling pathways such as Erk. Mnk-I1 or genetic knockout of the Mnks decreased the expression of vimentin, a marker of mesenchymal cells, without affecting vimentin mRNA levels. Vimentin protein levels were much lower in Mnk1/2-knockout cells than in controls, although mRNA levels were similar. Our data suggest that the Mnks regulate the translation of the vimentin mRNA and the stability of the vimentin protein. Inhibition or genetic knockout of the Mnks increased the binding of eIF4E to the cytoplasmic FMRP-interacting protein 1 (CYFIP1), which binds the fragile-X mental retardation protein, FMRP, a translational repressor. Since FMRP binds mRNAs for proteins involved in metastasis, the Mnk-dependent release of CYFIP1 from eIF4E is expected to release the repression of translation of FMRP-bound mRNAs, potentially providing a molecular mechanism for the control of cell migration by the Mnks. As Mnk1/2 are not essential for viability, inhibition of the Mnks may be a useful approach to tackling cancer metastasis, a key process contributing to mortality in cancer patients.
Collapse
|
39
|
Arnold A, Rahman MM, Lee MC, Muehlhaeusser S, Katic I, Gaidatzis D, Hess D, Scheckel C, Wright JE, Stetak A, Boag PR, Ciosk R. Functional characterization of C. elegans Y-box-binding proteins reveals tissue-specific functions and a critical role in the formation of polysomes. Nucleic Acids Res 2014; 42:13353-69. [PMID: 25378320 PMCID: PMC4245946 DOI: 10.1093/nar/gku1077] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The cold shock domain is one of the most highly conserved motifs between bacteria and higher eukaryotes. Y-box-binding proteins represent a subfamily of cold shock domain proteins with pleiotropic functions, ranging from transcription in the nucleus to translation in the cytoplasm. These proteins have been investigated in all major model organisms except Caenorhabditis elegans. In this study, we set out to fill this gap and present a functional characterization of CEYs, the C. elegans Y-box-binding proteins. We find that, similar to other organisms, CEYs are essential for proper gametogenesis. However, we also report a novel function of these proteins in the formation of large polysomes in the soma. In the absence of the somatic CEYs, polysomes are dramatically reduced with a simultaneous increase in monosomes and disomes, which, unexpectedly, has no obvious impact on animal biology. Because transcripts that are enriched in polysomes in wild-type animals tend to be less abundant in the absence of CEYs, our findings suggest that large polysomes might depend on transcript stabilization mediated by CEY proteins.
Collapse
Affiliation(s)
- Andreas Arnold
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland University of Basel, Petersplatz 1, CH-4003 Basel, Switzerland
| | - Md Masuder Rahman
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Man Chun Lee
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | | | - Iskra Katic
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland
| | - Dimos Gaidatzis
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland Swiss Institute of Bioinformatics, CH-4058 Basel, Switzerland
| | - Daniel Hess
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland
| | - Claudia Scheckel
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland University of Basel, Petersplatz 1, CH-4003 Basel, Switzerland
| | - Jane E Wright
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland
| | - Attila Stetak
- Department of Neuroscience, Biozentrum/Pharmazentrum, Basel 4046, Switzerland
| | - Peter R Boag
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Rafal Ciosk
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland
| |
Collapse
|
40
|
mTORC1 signaling controls multiple steps in ribosome biogenesis. Semin Cell Dev Biol 2014; 36:113-20. [PMID: 25148809 DOI: 10.1016/j.semcdb.2014.08.004] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/07/2014] [Accepted: 08/11/2014] [Indexed: 02/06/2023]
Abstract
Ribosome biogenesis is critical for cells to generate the ribosomes they need for protein synthesis in order to survive, grow and proliferate. It is a complex process, involving the coordinated production of four different RNA species and about 80 proteins, as well as their assembly into functional ribosomal subunits. Given its high demand for amino acids and nucleotides, it is also a metabolically expensive process for the cell. The mammalian target of rapamycin complex 1 (mTORC1) is a protein kinases which is activated by nutrients, anabolic hormones and oncogenic signaling pathways. mTORC1 positively regulates several steps in ribosome biogenesis, including ribosomal RNA transcription, the synthesis of ribosomal proteins and other components required for ribosome assembly. mTORC1 can thus coordinate stimuli which promote ribosome production with the various steps involved in this process. Although important advances have been made in our understanding of mTORC1 signaling, major questions remain about the molecular mechanisms by which it regulates ribosome biogenesis.
Collapse
|