1
|
Xiong X, Du Y, Liu P, Li X, Lai X, Miao H, Ning B. Unveiling EIF5A2: A multifaceted player in cellular regulation, tumorigenesis and drug resistance. Eur J Pharmacol 2025; 997:177596. [PMID: 40194645 DOI: 10.1016/j.ejphar.2025.177596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
The eukaryotic initiation factor 5A2 gene (EIF5A2) is a highly conserved and multifunctional gene that significantly influences various cellular processes, including translation elongation, RNA binding, ribosome binding, protein binding and post-translational modifications. Overexpression of EIF5A2 is frequently observed in multiple cancers, where it functions as an oncoprotein. Additionally, EIF5A2 is implicated in drug resistance through the regulation of various molecular pathways. In the review, we describe the structure and functions of EIF5A2 in normal cells and its role in tumorigenesis. We also elucidate the molecular mechanisms associated with EIF5A2 in the context of tumorigenesis and drug resistance. We propose that the biological roles of EIF5A2 in regulating diverse cellular processes and tumorigenesis are clinically significant and warrant further investigation.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China; Guangzhou Institute of Burn Clinical Medicine, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Yanli Du
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Peng Liu
- Departments of Burn and Plastic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xinye Li
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xudong Lai
- Department of infectious disease, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Haixiong Miao
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| | - Bo Ning
- Department of Neurosurgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| |
Collapse
|
2
|
Gao L, Tang Z, Zhang Z, Wei D, Wang J. Transcription factor FOS promotes ferroptosis and inflammation in S. aureus- infected osteomyelitis via EIF5A. J Orthop Surg Res 2025; 20:412. [PMID: 40270004 PMCID: PMC12020303 DOI: 10.1186/s13018-025-05815-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Osteomyelitis (OM) is a bone disease that can leave people disabled. Eukaryotic translation initiation factor (EIF5A) is involved in cell proliferation, apoptosis, differentiation, and inflammation, but the role of EIF5A in staphylococcus aureus (S. aureus)-infected OM remains unclear. METHODS The mRNA and proteins were detected by qRT-PCR and western blot. Cell viability was examined by CCK8 assay. The reactive oxygen species (ROS), malondialdehyde (MDA), ferrous iron (Fe2+), and glutathione (GSH) levels were analyzed using the ROS, MDA, GSH, and Fe2+ detection kits. The levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and Interleukin-6 (IL-6) were examined using Enzyme-linked immunosorbent (ELISA) kits. The binding between FOS and promoter of EIF5A was detected by chromatin immunoprecipitation (CHIP) assay. The interaction between EIF5A and Fos proto-oncogene (FOS) was detected by dual-luciferase reporter assay. The diagnostic values of EIF5A and FOS were analyzed with blood of S. aureus-infected OM patients and healthy volunteers by ROC curve. RESULTS The EIF5A was up-regulated in S. aureus-infected OM. EIF5A knockdown promoted cell viability in S. aureus-infected MG-63 cells and reduced ROS, MDA, and Fe2+ levels, and increased GSH levels. Meanwhile, silencing EIF5A could increase expression of glutathione peroxidase 4 (GPX4), and ferritin heavy chain1 (FTH1) and reduce acyl-CoA synthetase long-chain family member 4 (ACSL4) expression, and silencing EIF5A could reduce immune factors (TNF-α, IL-1β, and IL-6) levels. FOS could bind to EIF5A. Silencing FOS promoted cell viability, and increased GSH levels in S. aureus-infected MG-63 cells, but reduced ROS, MDA, and Fe2+ levels. Meanwhile, promoted GPX4 and FTH1 expression, inhibited ACSL4 expression, and reduced immune factor levels in S. aureus-infected MG-63 cells. Interestingly, EIF5A overexpression could weaken the actions. FOS promotes ferroptosis and inflammation via EIF5A in S. aureus-infected MG-63 cells. Besides, the EIF5A and FOS might be potential molecular diagnostic markers in the progression of OM. CONCLUSION FOS promotes ferroptosis and inflammation via EIF5A in S. aureus-infected OM. This study is first to report the role of FOS and EIF5A in S. aureus-infected OM, but we found that there are still some limitations in our work, such as not covering all possible types of infection, which is the focus of future research.
Collapse
Affiliation(s)
- Lei Gao
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China
| | - Zhipeng Tang
- Clinnical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Zhijin Zhang
- Department of Hand Surgery, Jiangnan Hospital Affiliated Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dehua Wei
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China
| | - Jiangning Wang
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China.
| |
Collapse
|
3
|
Guha A, Sadeghi SA, Kunhiraman HH, Fang F, Wang Q, Rafieioskouei A, Grumelot S, Gharibi H, Saei AA, Sayadi M, Weintraub NL, Horibata S, Yang PCM, Bonakdarpour B, Ghassemi M, Sun L, Mahmoudi M. AI-Driven Prediction of Cardio-Oncology Biomarkers Through Protein Corona Analysis. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2025; 509:161134. [PMID: 40190726 PMCID: PMC11970620 DOI: 10.1016/j.cej.2025.161134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Protein corona, a layer predominantly composed of proteins and other biomolecules that forms on nanoparticle surfaces upon interaction with biological fluids, has recently been extensively utilized to enhance the depth of plasma proteomics and biomarker discovery. In this study, we integrate protein corona profiling with mass spectrometry (MS)-based bottom-up proteomics (BUP), machine learning, and causality analysis to identify potential biomarkers in the field of cardio-oncology. We selected prostate cancer (PC) and atherosclerosis as model cardio-oncology diseases, given that PC is the most prevalent cancer among men in the United States and frequently coexists with atherosclerotic cardiovascular disease (ASCVD), which contributes to the progression of metastatic PC (mPC). Protein corona profiles were generated from 35 plasma samples categorized into four groups: mPC with ASCVD, nonmetastatic PC (nmPC) with ASCVD, mPC without ASCVD, and nmPC without ASCVD. MS-based BUP analysis identified 887 unique proteins within the protein corona. Gene Ontology (GO) analysis of the 260 proteins common to all samples revealed key plasma proteomic pathways significantly associated with ASCVD and mPC. Using Least Absolute Shrinkage and Selection Operator (LASSO) regularization, we isolated 22 proteins strongly associated with ASCVD or mPC, including chaperonin containing TCP1 subunit 7 (CCT7), which was common to both conditions. Automated formal reasoning and causality analysis of these 22 proteins identified thromboxane-A synthase 1 (TBXAS1) as a primary causal factor linked to both ASCVD and mPC. TBXAS1 plays a critical role in promoting platelet aggregation, vascular smooth muscle cell proliferation, endothelial dysfunction, and thrombosis. In this proof-of-concept study, CCT7 and TBXAS1 emerged as potential biomarkers for both ASCVD and mPC, suggesting their utility as dual biomarkers for early detection and targeted therapeutic interventions. By combining nanomedicine with advanced analytical methods, our integrated approach provides a robust framework for uncovering causal relationships between biomarkers and disease states.
Collapse
Affiliation(s)
- Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | | | - Harikrishnan Hyma Kunhiraman
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fei Fang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Qianyi Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Arshia Rafieioskouei
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Shaun Grumelot
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Hassan Gharibi
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Amir Ata Saei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maryam Sayadi
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Neal L. Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Sachi Horibata
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Phillip Chung-Ming Yang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Borzoo Bonakdarpour
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Mohammad Ghassemi
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
4
|
Benaceur O, Ferreira Montenegro P, Kahi M, Fontaine-Vive F, Mazure NM, Mehiri M, Bost F, Peraldi P. Development of a reliable, sensitive, and convenient assay for the discovery of new eIF5A hypusination inhibitors. PLoS One 2025; 20:e0308049. [PMID: 39937781 DOI: 10.1371/journal.pone.0308049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/21/2024] [Indexed: 02/14/2025] Open
Abstract
eIF5A is a translation factor dysregulated in several pathologies such as cancer and diabetes. eIF5A activity depends upon its hypusination, a unique post-translational modification catalyzed by two enzymes: DHPS and DOHH. Only a few molecules able to inhibit hypusination have been described, and none are used for the treatment of patients. The scarcity of new inhibitors is probably due to the challenge of measuring DHPS and DOHH activities. Here, we describe the Hyp'Assay, a convenient cell-free assay to monitor eIF5A hypusination. Hypusination is performed in 96-well plates using recombinant human eIF5A, DHPS, and DOHH and is revealed by an antibody against hypusinated eIF5A. Pharmacological values obtained with the Hyp'Assay, such as the EC50 of DHPS for spermidine or the IC50 of GC7 for DHPS, were similar to published data, supporting the reliability of the Hyp'Assay. As a proof of concept, we synthesized four new GC7 analogs and showed, using the Hyp'Assay, that these derivatives inhibit hypusination. In summary, we present the Hyp'Assay; a reliable and sensitive assay for new hypusination inhibitors. This assay could be of interest to researchers wanting an easier way to study hypusination, and also a valuable tool for large-scale screening of chemical libraries for new hypusination inhibitors.
Collapse
Affiliation(s)
| | | | - Michel Kahi
- Université Côte d'Azur, Inserm U1065, C3M, Nice, France
| | | | | | - Mohamed Mehiri
- Université Côte d'Azur, CNRS UMR 7272, Institut de Chimie de Nice, Nice, France
| | - Frederic Bost
- Université Côte d'Azur, Inserm U1065, C3M, Nice, France
| | | |
Collapse
|
5
|
Schneider-Poetsch T, Dang Y, Iwasaki W, Arata M, Shichino Y, Al Mourabit A, Moriou C, Romo D, Liu JO, Ito T, Iwasaki S, Yoshida M. Girolline is a sequence context-selective modulator of eIF5A activity. Nat Commun 2025; 16:223. [PMID: 39794322 PMCID: PMC11724050 DOI: 10.1038/s41467-024-54838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/21/2024] [Indexed: 01/13/2025] Open
Abstract
Natural products have a long history of providing probes into protein biosynthesis, with many of these compounds serving as therapeutics. The marine natural product girolline has been described as an inhibitor of protein synthesis. Its precise mechanism of action, however, has remained unknown. The data we present here suggests that girolline is a sequence-selective modulator of translation factor eIF5A. Girolline interferes with ribosome-eIF5A interaction and induces ribosome stalling where translational progress is impeded, including on AAA-encoded lysine. Our data furthermore indicate that eIF5A plays a physiological role in ribosome-associated quality control and in maintaining the efficiency of translational progress. Girolline helped to deepen our understanding of the interplay between protein production and quality control in a physiological setting and offers a potent chemical tool to selectively modulate gene expression.
Collapse
Affiliation(s)
- Tilman Schneider-Poetsch
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan.
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Wakana Iwasaki
- Laboratory for Translation Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Mayumi Arata
- Drug Discovery Seed Compounds Exploratory Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan
| | - Ali Al Mourabit
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Celine Moriou
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, Waco, USA
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takuhiro Ito
- Laboratory for Translation Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Shintaro Iwasaki
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Minoru Yoshida
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan.
- Drug Discovery Seed Compounds Exploratory Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan.
- Office of University Professors, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
6
|
Kumar D, Kanchan R, Chaturvedi NK. Targeting protein synthesis pathways in MYC-amplified medulloblastoma. Discov Oncol 2025; 16:23. [PMID: 39779613 PMCID: PMC11711608 DOI: 10.1007/s12672-025-01761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
MYC is one of the most deregulated oncogenic transcription factors in human cancers. MYC amplification/or overexpression is most common in Group 3 medulloblastoma and is positively associated with poor prognosis. MYC is known to regulate the transcription of major components of protein synthesis (translation) machinery, leading to promoted rates of protein synthesis and tumorigenesis. MTOR signaling-driven deregulated protein synthesis is widespread in various cancers, including medulloblastoma, which can promote the stabilization of MYC. Indeed, our previous studies demonstrate that the key components of protein synthesis machinery, including mTOR signaling and MYC targets, are overexpressed and activated in MYC-amplified medulloblastoma, confirming MYC-dependent addiction of enhanced protein synthesis in medulloblastoma. Further, targeting this enhanced protein synthesis pathway with combined inhibition of MYC transcription and mTOR translation by small-molecule inhibitors, demonstrates preclinical synergistic anti-tumor potential against MYC-driven medulloblastoma in vitro and in vivo. Thus, inhibiting enhanced protein synthesis by targeting the MYC indirectly and mTOR pathways together may present a highly appropriate strategy for treating MYC-driven medulloblastoma and other MYC-addicted cancers. Evidence strongly proposes that MYC/mTOR-driven tumorigenic signaling can predominantly control the translational machinery to elicit cooperative effects on increased cell proliferation, cell cycle progression, and genome dysregulation as a mechanism of cancer initiation. Several small molecule inhibitors of targeting MYC indirectly and mTOR signaling have been developed and used clinically with immunosuppressants and chemotherapy in multiple cancers. Only a few of them have been investigated as treatments for medulloblastoma and other pediatric tumors. This review explores concurrent targeting of MYC and mTOR signaling against MYC-driven medulloblastoma. Based on existing evidence, targeting of MYC and mTOR pathways together produces functional synergy that could be the basis for effective therapies against medulloblastoma.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA
| | - Ranjana Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA.
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
7
|
Zhao G, Zhao X, Liu Z, Wang B, Dong P, Watari H, Pfeffer LM, Tigyi G, Zhang W, Yue J. Knockout or inhibition of DHPS suppresses ovarian tumor growth and metastasis by attenuating the TGFβ pathway. Sci Rep 2025; 15:917. [PMID: 39762448 PMCID: PMC11704301 DOI: 10.1038/s41598-025-85466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Deoxyhypusine synthase (DHPS) is an enzyme encoded by the DHPS gene, with high expression in various cancers, including ovarian cancer (OC). DHPS regulates the translation initiation factor EIF5A, and EIF5A2 knockout inhibits OC tumor growth and metastasis by blocking the epithelial-to-mesenchymal transition (EMT) and the TGFβ pathway. In this study, we show that DHPS is amplified in OC patients, and its elevated expression correlates with poor survival. Using lentiviral CRISPR/Cas9 vectors for DHPS knockout, we observed EMT inhibition in SKOV3 and OVCAR8 cells through suppressed hypusination and reduced EIF5A2 expression. Inhibition of DHPS activity with GC7 similarly blocked hypusination and EMT. Disrupting DHPS expression, either genetically or pharmacologically, inhibited primary tumor growth and metastasis in OC mouse models. These findings suggest that targeting DHPS and inhibiting hypusination could be promising strategies for OC treatment.
Collapse
Affiliation(s)
- Guannan Zhao
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Xinxin Zhao
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Ziping Liu
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Baojin Wang
- Department of Obstetrics and Gynecology, The Third Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Gabor Tigyi
- Department of Physiology, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, Collage of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, United States.
- Center for Cancer Research, Collage of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
8
|
Guo K, Zhou J. Insights into eukaryotic translation initiation factor 5A: Its role and mechanisms in protein synthesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119849. [PMID: 39303786 DOI: 10.1016/j.bbamcr.2024.119849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
The protein synthesis within eukaryotic cells is a complex process involving various translation factors. Among these factors, eukaryotic translation initiation factor 5 A (eIF5A) emerges as a crucial translation factor with high evolutionary conservation. eIF5A is unique as it is the only protein in eukaryotic cells containing the hypusine modification. Initially presumed to be a translation initiation factor, eIF5A was subsequently discovered to act mainly during the translation elongation phase. Notably, eIF5A facilitates the translation of peptide sequences containing polyproline stretches and exerts a universal regulatory effect on the elongation and termination phases of protein synthesis. Additionally, eIF5A indirectly affects various physiological processes within the cell by modulating the translation of specific proteins. This review provides a comprehensive overview of the structure, physiological functions, various post-translational modifications of eIF5A, and its association with various human diseases. The comparison between eIF5A and its bacterial homolog, EF-P, extends the discussion to the evolutionary conservation of eIF5A. This highlights its significance across different domains of life.
Collapse
Affiliation(s)
- Keying Guo
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
9
|
Thangavelu L, Imran M, Alsharari SH, Abdulaziz AM, Alawlaqi AM, Kamal M, Rekha MM, Kaur M, Soothwal P, Arora I, Kumar MR, Chauhan AS. Exploring hypoxia-induced ncRNAs as biomarkers and therapeutic targets in lung cancer. Pathol Res Pract 2024; 263:155613. [PMID: 39383737 DOI: 10.1016/j.prp.2024.155613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024]
Abstract
Lung cancer is a deadly disease, causing nearly 20 % of all cancer deaths globally. A key factor in lung cancer's development and resistance to treatment is hypoxia, a condition where tumor cells experience low oxygen levels. In this low-oxygen environment, special molecules called non-coding RNAs (ncRNAs) become critical players. NcRNAs, including lncRNAs, miRNAs, circRNAs, and siRNAs, control how genes function and how cells behave. Some ncRNAs, like HIF1A-AS2 and HOTAIR, are linked to the aggressive spread of lung cancer, making them potential targets for therapy. Others, like certain miRNAs, show promise as early detection tools due to their influence on tumor blood vessel formation and metabolism. This complex interplay between hypoxia and ncRNAs is crucial for understanding lung cancer. For example, circRNAs can control the activity of miRNAs, impacting how tumors respond to low oxygen. Additionally, siRNAs offer a potential strategy to overcome treatment resistance caused by hypoxia. By studying the intricate relationship between hypoxia and ncRNAs, scientists hope to uncover new biomarkers for lung cancer. This knowledge will pave the way for developing more effective and targeted treatments for this devastating disease.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | | | - Akrm M Abdulaziz
- Department of Clinical Pharmacy, King Khalid Hospital, Najran 66262, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pradeep Soothwal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Isha Arora
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India.
| |
Collapse
|
10
|
D'Agostino M, Simonetti A, Motta S, Wolff P, Romagnoli A, Piccinini A, Spinozzi F, Di Marino D, La Teana A, Ennifar E. Crystal structure of archaeal IF5A-DHS complex reveals insights into the hypusination mechanism. Structure 2024; 32:878-888.e4. [PMID: 38582076 DOI: 10.1016/j.str.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/12/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
The translation factor IF5A is highly conserved in Eukarya and Archaea and undergoes a unique post-translational hypusine modification by the deoxyhypusine synthase (DHS) enzyme. DHS transfers the butylamine moiety from spermidine to IF5A using NAD as a cofactor, forming a deoxyhypusine intermediate. IF5A is a key player in protein synthesis, preventing ribosome stalling in proline-rich sequences during translation elongation and facilitating translation elongation and termination. Additionally, human eIF5A participates in various essential cellular processes and contributes to cancer metastasis, with inhibiting hypusination showing anti-proliferative effects. The hypusination pathway of IF5A is therefore an attractive new therapeutic target. We elucidated the 2.0 Å X-ray crystal structure of the archaeal DHS-IF5A complex, revealing hetero-octameric architecture and providing a detailed view of the complex active site including the hypusination loop. This structure, along with biophysical data and molecular dynamics simulations, provides new insights into the catalytic mechanism of the hypusination reaction.
Collapse
Affiliation(s)
- Mattia D'Agostino
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; Architecture et Réactivité de l'ARN, CNRS UPR 9002, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Angelita Simonetti
- Architecture et Réactivité de l'ARN, CNRS UPR 9002, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| | - Philippe Wolff
- Architecture et Réactivité de l'ARN, CNRS UPR 9002, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Alice Romagnoli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; New York-Marche Structural Biology Center (Ny-Masbic), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Astra Piccinini
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Francesco Spinozzi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; New York-Marche Structural Biology Center (Ny-Masbic), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Via Mario Negri 2, 20156 Milano, Italy.
| | - Anna La Teana
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; New York-Marche Structural Biology Center (Ny-Masbic), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Eric Ennifar
- Architecture et Réactivité de l'ARN, CNRS UPR 9002, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
11
|
Lou D, Jia Q, Zhang H, Wang J, Liu L, Liu Z, Jia X, Wang J, Shan C. MiR-5189-3p Suppresses cell Proliferation, Invasion and Migration Through Targeting EIF5A2 in Laryngeal Squamous Cell Carcinoma. Biochem Genet 2024; 62:1603-1616. [PMID: 37656330 DOI: 10.1007/s10528-023-10489-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/06/2023] [Indexed: 09/02/2023]
Abstract
A growing body of evidence suggests that miR-5189-3p plays a critical role in multiple diseases. This study aimed to investigate the function of miR-5189-3p in laryngeal squamous cell carcinoma (LSCC) and explore its underlying mechanisms. qRT-PCR was designed to determine the expression levels of miR-5189-3p and eukaryotic translation initiation factor 5A2 (EIF5A2), while CCK-8 assay was performed to measure the effects of miR-5189-3p on cell proliferation. Transwell assay was performed to evaluate cell invasion as well as migration, and wound healing assay was applied to demonstrate cell migratory ability. Target gene prediction and luciferase reporter assay were developed to screen the possible target gene of miR-5189-3p, and Western blot was designed to measure EIF5A2 protein expression. MiR-5189-3p was down-regulated in LSCC tissues and cell lines. Up-regulation of miR-5189-3p notably inhibited cell proliferation, invasion, and migration in HEP2 and FADU cells. EIF5A2 was the potential downstream gene of miR-5189-3p, and overexpression of miR-5189-3p apparently reduced EIF5A2 expression. Moreover, reintroduction of EIF5A2 rescued the tumor suppressive effects of miR-5189-3p. MiR-5189-3p functions as a tumor inhibitor in LSCC progression via directly regulating EIF5A2 and may be a potential therapeutic target for LSCC.
Collapse
Affiliation(s)
- Dan Lou
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
- Department of Otolaryngology, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Qiaojing Jia
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Haizhong Zhang
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Jingmiao Wang
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Lisha Liu
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Zhichang Liu
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaofang Jia
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Jianxing Wang
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| | - Chunguang Shan
- Department of Otolaryngology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
12
|
Martin-Solana E, Diaz-Lopez I, Mohamedi Y, Ventoso I, Fernandez JJ, Fernandez-Fernandez MR. Progressive alterations in polysomal architecture and activation of ribosome stalling relief factors in a mouse model of Huntington's disease. Neurobiol Dis 2024; 195:106488. [PMID: 38565397 PMCID: PMC7616275 DOI: 10.1016/j.nbd.2024.106488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Given their highly polarized morphology and functional singularity, neurons require precise spatial and temporal control of protein synthesis. Alterations in protein translation have been implicated in the development and progression of a wide range of neurological and neurodegenerative disorders, including Huntington's disease (HD). In this study we examined the architecture of polysomes in their native brain context in striatal tissue from the zQ175 knock-in mouse model of HD. We performed 3D electron tomography of high-pressure frozen and freeze-substituted striatal tissue from HD models and corresponding controls at different ages. Electron tomography results revealed progressive remodelling towards a more compacted polysomal architecture in the mouse model, an effect that coincided with the emergence and progression of HD related symptoms. The aberrant polysomal architecture is compatible with ribosome stalling phenomena. In fact, we also detected in the zQ175 model an increase in the striatal expression of the stalling relief factor EIF5A2 and an increase in the accumulation of eIF5A1, eIF5A2 and hypusinated eIF5A1, the active form of eIF5A1. Polysomal sedimentation gradients showed differences in the relative accumulation of 40S ribosomal subunits and in polysomal distribution in striatal samples of the zQ175 model. These findings indicate that changes in the architecture of the protein synthesis machinery may underlie translational alterations associated with HD, opening new avenues for understanding the progression of the disease.
Collapse
Affiliation(s)
- Eva Martin-Solana
- Centro Nacional de Biotecnología (CNB-CSIC). Campus UAM, Darwin 3, 28049 Madrid, Spain
| | - Irene Diaz-Lopez
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Yamina Mohamedi
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA). Av. Hospital Universitario s/n, 33011 Oviedo, Asturias, Spain
| | - Ivan Ventoso
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM) and Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jose-Jesus Fernandez
- Centro Nacional de Biotecnología (CNB-CSIC). Campus UAM, Darwin 3, 28049 Madrid, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA). Av. Hospital Universitario s/n, 33011 Oviedo, Asturias, Spain; Centro de Investigación en Nanomateriales y Nanotecnología (CINN-CSIC). Av. Vega 4-6, 33940 El Entrego, Asturias, Spain.
| | - Maria Rosario Fernandez-Fernandez
- Centro Nacional de Biotecnología (CNB-CSIC). Campus UAM, Darwin 3, 28049 Madrid, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA). Av. Hospital Universitario s/n, 33011 Oviedo, Asturias, Spain; Centro de Investigación en Nanomateriales y Nanotecnología (CINN-CSIC). Av. Vega 4-6, 33940 El Entrego, Asturias, Spain.
| |
Collapse
|
13
|
Francis JC, Capper A, Rust AG, Ferro K, Ning J, Yuan W, de Bono J, Pettitt SJ, Swain A. Identification of genes that promote PI3K pathway activation and prostate tumour formation. Oncogene 2024; 43:1824-1835. [PMID: 38654106 PMCID: PMC11164682 DOI: 10.1038/s41388-024-03028-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
We have performed a functional in vivo mutagenesis screen to identify genes that, when altered, cooperate with a heterozygous Pten mutation to promote prostate tumour formation. Two genes, Bzw2 and Eif5a2, which have been implicated in the process of protein translation, were selected for further validation. Using prostate organoid models, we show that either Bzw2 downregulation or EIF5A2 overexpression leads to increased organoid size and in vivo prostate growth. We show that both genes impact the PI3K pathway and drive a sustained increase in phospho-AKT expression, with PTEN protein levels reduced in both models. Mechanistic studies reveal that EIF5A2 is directly implicated in PTEN protein translation. Analysis of patient datasets identified EIF5A2 amplifications in many types of human cancer, including the prostate. Human prostate cancer samples in two independent cohorts showed a correlation between increased levels of EIF5A2 and upregulation of a PI3K pathway gene signature. Consistent with this, organoids with high levels of EIF5A2 were sensitive to AKT inhibitors. Our study identified novel genes that promote prostate cancer formation through upregulation of the PI3K pathway, predicting a strategy to treat patients with genetic aberrations in these genes particularly relevant for EIF5A2 amplified tumours.
Collapse
Affiliation(s)
- Jeffrey C Francis
- Division of Cancer Biology, Institute of Cancer Research, London, SW3 6JB, UK
| | - Amy Capper
- Division of Cancer Biology, Institute of Cancer Research, London, SW3 6JB, UK
| | - Alistair G Rust
- Genomics Facility, Institute of Cancer Research, London, UK
- Genomic Data Sciences, GlaxoSmithKline, Stevenage, UK
| | - Klea Ferro
- Division of Cancer Biology, Institute of Cancer Research, London, SW3 6JB, UK
| | - Jian Ning
- Tumour Modelling Facility, Institute of Cancer Research, London, SW3 6JB, UK
| | - Wei Yuan
- Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - Johann de Bono
- Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - Stephen J Pettitt
- The CRUK Gene Function Laboratory, Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | - Amanda Swain
- Division of Cancer Biology, Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
14
|
Abdel-Azim G, Patel P, Li S, Guo S, Black MH. Fast multiple-trait genome-wide association analysis for correlated longitudinal measurements. Sci Rep 2023; 13:20603. [PMID: 37996550 PMCID: PMC10667366 DOI: 10.1038/s41598-023-47555-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Large-scale longitudinal biobank data can be leveraged to identify genetic variation contributing to human diseases progression and traits trajectories. While methods for genome-wide association studies (GWAS) of multiple correlated traits have been proposed, an efficient multiple-trait approach to model longitudinal phenotypes is not currently available. We developed GAMUT, a genome-wide association approach for multiple longitudinal traits. GAMUT employs a mixed-effects model to fit longitudinal outcomes where a fast algorithm for inversion by recursive partitioning of the random effects submatrix is introduced. To evaluate performance of the algorithms introduced and assess their statistical power and type I error, stochastic simulation was conducted. Consistent with our expectation, power was greater for cross-sectional (CS) than longitudinal (LT) effects, particularly with a diminishing LT/CS ratio. With a minimum minor allele count of 3 within genotype by time categories, observed type I error was roughly equal to theoretical genome-wide significance. Additionally, 28 blood-based biomarkers measured at 2 time points on participants of the UK Biobank were used to compare GAMUT against single-trait standard and longitudinal GWAS (including rate of change). Across all biomarkers, we observed 539 (CS) and 248 (LT) significant independent variants for the GAMUT method, and 513 (CS) and 30 (LT) for single-trait longitudinal GWAS, respectively. Only 37 variants were identified by modeling rates of change using standard GWAS.
Collapse
Affiliation(s)
| | - Parth Patel
- Janssen Res. & Dev. (Johnson & Johnson), Spring House, PA, USA
| | - Shuwei Li
- Janssen Res. & Dev. (Johnson & Johnson), Spring House, PA, USA
| | - Shicheng Guo
- Janssen Res. & Dev. (Johnson & Johnson), Spring House, PA, USA
| | | |
Collapse
|
15
|
Guo JS, Liu KL, Qin YX, Hou L, Jian LY, Yang YH, Li XY. Hypusination-induced DHPS/eIF5A pathway as a new therapeutic strategy for human diseases: A mechanistic review and structural classification of DHPS inhibitors. Biomed Pharmacother 2023; 167:115440. [PMID: 37683595 DOI: 10.1016/j.biopha.2023.115440] [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: 07/04/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
The discovery of new therapeutic strategies for diseases is essential for drug research. Deoxyhypusine synthase (DHPS) is a critical enzyme that modifies the conversion of the eukaryotic translation initiation factor 5A (eIF5A) precursor into physiologically active eIF5A (eIF5A-Hyp). Recent studies have revealed that the hypusine modifying of DHPS on eIF5A has an essential regulatory role in human diseases. The hypusination-induced DHPS/eIF5A pathway has been shown to play an essential role in various cancers, and it could regulate immune-related diseases, glucose metabolism-related diseases, neurological-related diseases, and aging. In addition, DHPS has a more defined substrate and a well-defined structure within the active pocket than eIF5A. More and more researchers are focusing on the prospect of advanced development of DHPS inhibitors. This review summarizes the regulatory mechanisms of the hypusination-induced DHPS/eIF5A pathway in a variety of diseases in addition to the inhibitors related to this pathway; it highlights and analyzes the structural features and mechanisms of action of DHPS inhibitors and expands the prospects of future drug development using DHPS as an anticancer target.
Collapse
Affiliation(s)
- Jing-Si Guo
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, No. 77 Puhe, Shenyang 110122, PR China
| | - Yu-Xi Qin
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China
| | - Lin Hou
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China
| | - Ling-Yan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China
| | - Yue-Hui Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China
| | - Xin-Yang Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, PR China.
| |
Collapse
|
16
|
Padgett LR, Shinkle MR, Rosario S, Stewart TM, Foley JR, Casero RA, Park MH, Chung WK, Mastracci TL. Deoxyhypusine synthase mutations alter the post-translational modification of eukaryotic initiation factor 5A resulting in impaired human and mouse neural homeostasis. HGG ADVANCES 2023; 4:100206. [PMID: 37333770 PMCID: PMC10275725 DOI: 10.1016/j.xhgg.2023.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
DHPS deficiency is a rare genetic disease caused by biallelic hypomorphic variants in the Deoxyhypusine synthase (DHPS) gene. The DHPS enzyme functions in mRNA translation by catalyzing the post-translational modification, and therefore activation, of eukaryotic initiation factor 5A (eIF5A). The observed clinical outcomes associated with human mutations in DHPS include developmental delay, intellectual disability, and seizures. Therefore, to increase our understanding of this rare disease, it is critical to determine the mechanisms by which mutations in DHPS alter neurodevelopment. In this study, we have generated patient-derived lymphoblast cell lines and demonstrated that human DHPS variants alter DHPS protein abundance and impair enzyme function. Moreover, we observe a shift in the abundance of the post-translationally modified forms of eIF5A; specifically, an increase in the nuclear localized acetylated form (eIF5AAcK47) and concomitant decrease in the cytoplasmic localized hypusinated form (eIF5AHYP). Generation and characterization of a mouse model with a genetic deletion of Dhps in the brain at birth shows that loss of hypusine biosynthesis impacts neuronal function due to impaired eIF5AHYP-dependent mRNA translation; this translation defect results in altered expression of proteins required for proper neuronal development and function. This study reveals new insight into the biological consequences and molecular impact of human DHPS deficiency and provides valuable information toward the goal of developing treatment strategies for this rare disease.
Collapse
Affiliation(s)
- Leah R. Padgett
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Mollie R. Shinkle
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jackson R. Foley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert A. Casero
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Myung Hee Park
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4340, USA
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - Teresa L. Mastracci
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
17
|
Wątor E, Wilk P, Biela A, Rawski M, Zak KM, Steinchen W, Bange G, Glatt S, Grudnik P. Cryo-EM structure of human eIF5A-DHS complex reveals the molecular basis of hypusination-associated neurodegenerative disorders. Nat Commun 2023; 14:1698. [PMID: 36973244 PMCID: PMC10042821 DOI: 10.1038/s41467-023-37305-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Hypusination is a unique post-translational modification of the eukaryotic translation factor 5A (eIF5A) that is essential for overcoming ribosome stalling at polyproline sequence stretches. The initial step of hypusination, the formation of deoxyhypusine, is catalyzed by deoxyhypusine synthase (DHS), however, the molecular details of the DHS-mediated reaction remained elusive. Recently, patient-derived variants of DHS and eIF5A have been linked to rare neurodevelopmental disorders. Here, we present the cryo-EM structure of the human eIF5A-DHS complex at 2.8 Å resolution and a crystal structure of DHS trapped in the key reaction transition state. Furthermore, we show that disease-associated DHS variants influence the complex formation and hypusination efficiency. Hence, our work dissects the molecular details of the deoxyhypusine synthesis reaction and reveals how clinically-relevant mutations affect this crucial cellular process.
Collapse
Affiliation(s)
- Elżbieta Wątor
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Piotr Wilk
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Artur Biela
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michał Rawski
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof M Zak
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Wieland Steinchen
- Philipps-University Marburg, Center for Synthetic Microbiology (SYNMIKRO) & Faculty of Chemistry, Marburg, Germany
| | - Gert Bange
- Philipps-University Marburg, Center for Synthetic Microbiology (SYNMIKRO) & Faculty of Chemistry, Marburg, Germany
- Max Planck Institute for Terrestrial Microbiology, Molecular Physiology of Microbes, Marburg, Germany
| | - Sebastian Glatt
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Przemysław Grudnik
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
18
|
Wu YY, Wu GQ, Cai NL, Xu YM, Lau ATY. Comparison of Human Eukaryotic Translation Initiation Factors 5A1 and 5AL1: Identification of Amino Acid Residues Important for EIF5A1 Lysine 50 Hypusination and Its Protein Stability. Int J Mol Sci 2023; 24:ijms24076067. [PMID: 37047039 PMCID: PMC10093921 DOI: 10.3390/ijms24076067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 04/14/2023] Open
Abstract
The human eukaryotic translation initiation factor 5A (EIF5A) family consists of three members, namely EIF5A1, EIF5A2, and EIF5AL1. Recent studies have shown that the expression of EIF5As is related to many human diseases, such as diabetes, viral infection, central nervous system injury, and cancer. Among them, EIF5A1 plays different functions in various cancers, possibly as a tumor-suppressor or oncogene, while EIF5A2 promotes the occurrence and development of cancer. Yet, the biological function of EIF5AL1 is not being studied so far. Interestingly, although there are only three amino acid (at residues 36, 45, and 109) differences between EIF5A1 and EIF5AL1, we demonstrate that only EIF5A1 can be hypusinated while EIF5AL1 cannot, and EIF5AL1 has a tumor-suppressor-like function by inhibiting cell proliferation and migration. We also show that EIF5AL1 protein turnover is mediated through the proteasomal pathway, and EIF5AL1 protein turnover is much faster than that of EIF5A1, which may explain their differential protein expression level in cells. By engineering single and double mutations on these three amino acids, we pinpoint which of these amino acids are critical for hypusination and protein stability. The data of this work should fill in the gaps in EIF5As research and pave the way for future studies on EIF5AL1.
Collapse
Affiliation(s)
- Yu-Yao Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Gao-Qi Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Na-Li Cai
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
19
|
Chen Y, León-Letelier RA, Abdel Sater AH, Vykoukal J, Dennison JB, Hanash S, Fahrmann JF. c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy. Cancers (Basel) 2023; 15:623. [PMID: 36765581 PMCID: PMC9913358 DOI: 10.3390/cancers15030623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
c-MYC and its paralogues MYCN and MYCL are among the most frequently amplified and/or overexpressed oncoproteins in ovarian cancer. c-MYC plays a key role in promoting ovarian cancer initiation and progression. The polyamine pathway is a bona fide target of c-MYC signaling, and polyamine metabolism is strongly intertwined with ovarian malignancy. Targeting of the polyamine pathway via small molecule inhibitors has garnered considerable attention as a therapeutic strategy for ovarian cancer. Herein, we discuss the involvement of c-MYC signaling and that of its paralogues in promoting ovarian cancer tumorigenesis. We highlight the potential of targeting c-MYC-driven polyamine metabolism for the treatment of ovarian cancers and the utility of polyamine signatures in biofluids for early detection applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
20
|
Mudryi V, Peske F, Rodnina M. Translation Factor Accelerating Peptide Bond Formation on the Ribosome: EF-P and eIF5A as Entropic Catalysts and a Potential Drug Targets. BBA ADVANCES 2023; 3:100074. [PMID: 37082265 PMCID: PMC10074943 DOI: 10.1016/j.bbadva.2023.100074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023] Open
Abstract
Elongation factor P (EF-P) and its eukaryotic homolog eIF5A are auxiliary translation factors that facilitate peptide bond formation when several sequential proline (Pro) residues are incorporated into the nascent chain. EF-P and eIF5A bind to the exit (E) site of the ribosome and contribute to favorable entropy of the reaction by stabilizing tRNA binding in the peptidyl transferase center of the ribosome. In most organisms, EF-P and eIF5A carry a posttranslational modification that is crucial for catalysis. The chemical nature of the modification varies between different groups of bacteria and between pro- and eukaryotes, making the EF-P-modification enzymes promising targets for antibiotic development. In this review, we summarize our knowledge of the structure and function of EF-P and eIF5A, describe their modification enzymes, and present an approach for potential drug screening aimed at EarP, an enzyme that is essential for EF-P modification in several pathogenic bacteria.
Collapse
|
21
|
Sfakianos AP, Raven RM, Willis AE. The pleiotropic roles of eIF5A in cellular life and its therapeutic potential in cancer. Biochem Soc Trans 2022; 50:1885-1895. [PMID: 36511302 PMCID: PMC9788402 DOI: 10.1042/bst20221035] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 11/19/2023]
Abstract
Protein synthesis is dysregulated in the majority of cancers and this process therefore provides a good therapeutic target. Many novel anti-cancer agents are directed to target the initiation stage of translation, however, translation elongation also holds great potential as a therapeutic target. The elongation factor eIF5A that assists the formation of peptidyl bonds during the elongation process is of considerable interest in this regard. Overexpression of eIF5A has been linked with the development of a variety of cancers and inhibitors of the molecule have been proposed for anti-cancer clinical applications. eIF5A is the only protein in the cell that contains the post-translational modification hypusine. Hypusination is a two-step enzymatic process catalysed by the Deoxyhypusine Synthase (DHPS) and Deoxyhypusine Hydroxylase (DOHH). In addition, eIF5A can be acetylated by p300/CBP-associated factor (PCAF) which leads to translocation of the protein to the nucleus and its deactivation. In addition to the nucleus, eIF5A has been found in the mitochondria and the endoplasmic reticulum (ER) with eIF5A localisation related to function from regulation of mitochondrial activity and apoptosis to maintenance of ER integrity and control of the unfolded protein response (UPR). Given the pleiotropic functions of eIF5A and by extension the hypusination enzymes, this system is being considered as a target for a range of cancers including multiple myeloma, B-Cell lymphoma, and neuroblastoma. In this review, we explore the role of eIF5A and discuss the therapeutic strategies that are currently developing both in the pre- and the clinical stage.
Collapse
Affiliation(s)
| | - Rebecca Mallory Raven
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge, U.K
| | - Anne Elizabeth Willis
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Rd, Cambridge, U.K
| |
Collapse
|
22
|
Farache D, Antine SP, Lee ASY. Moonlighting translation factors: multifunctionality drives diverse gene regulation. Trends Cell Biol 2022; 32:762-772. [PMID: 35466028 PMCID: PMC9378348 DOI: 10.1016/j.tcb.2022.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/09/2022]
Abstract
Translation factors have traditionally been viewed as proteins that drive ribosome function and ensure accurate mRNA translation. Recent discoveries have highlighted that these factors can also moonlight in gene regulation, but through functions distinct from their canonical roles in protein synthesis. Notably, the additional functions that translation factors encode are diverse, ranging from transcriptional control and extracellular signaling to RNA binding, and are highly regulated in response to external cues and the intrinsic cellular state. Thus, this multifunctionality of translation factors provides an additional mechanism for exquisite control of gene expression.
Collapse
Affiliation(s)
- Dorian Farache
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sadie P Antine
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy S Y Lee
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
23
|
Quantitative proteomics to study aging in rabbit spleen tissues. Exp Gerontol 2022; 167:111908. [PMID: 35932934 DOI: 10.1016/j.exger.2022.111908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/04/2022]
Abstract
Aging is a process that occurs in tissues and across species, leading to the degradation of many biological processes. We previously demonstrated that rabbits are a feasible model for studying aging due to their genetic homology and relatively short lifespan in comparison to humans. We utilized a cPILOT multiplexing strategy to identify proteomic changes in spleen tissues of young, middle, and old aged rabbits. We identified 63 proteins that change significantly (p < 0.05) with age and notably these proteins relate to nucleotide and RNA binding, DNA repair, actin regulation, and immune system pathways. Here, we explore the implications of aging in the spleen and demonstrate the utility of a rabbit model to understand aging processes.
Collapse
|
24
|
Farache D, Liu L, Lee ASY. Eukaryotic Initiation Factor 5A2 Regulates Expression of Antiviral Genes. J Mol Biol 2022; 434:167564. [PMID: 35358571 PMCID: PMC11906106 DOI: 10.1016/j.jmb.2022.167564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 12/14/2022]
Abstract
Translation factors are essential for regulation of protein synthesis. The eukaryotic translation initiation factor 5A (eIF5A) family is made up of two paralogues - eIF5A1 and eIF5A2 - which display high sequence homology but distinct tissue tropism. While eIF5A1 directly binds to the ribosome and regulates translation initiation, elongation, and termination, the molecular function of eIF5A2 remains poorly understood. Here, we engineer an eIF5A2 knockout allele in the SW480 colon cancer cell line. Using ribosome profiling and RNA-Sequencing, we reveal that eIF5A2 is functionally distinct from eIF5A1 and does not regulate transcript-specific or global protein synthesis. Instead, eIF5A2 knockout leads to decreased intrinsic antiviral gene expression, including members of the IFITM and APOBEC3 family. Furthermore, cells lacking eIF5A2 display increased permissiveness to virus infection. Our results uncover eIF5A2 as a factor involved regulating the antiviral transcriptome, and reveal an example of how gene duplications of translation factors can result in proteins with distinct functions.
Collapse
Affiliation(s)
- Dorian Farache
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Luochen Liu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Amy S Y Lee
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Kovalski JR, Kuzuoglu‐Ozturk D, Ruggero D. Protein synthesis control in cancer: selectivity and therapeutic targeting. EMBO J 2022; 41:e109823. [PMID: 35315941 PMCID: PMC9016353 DOI: 10.15252/embj.2021109823] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 11/09/2022] Open
Abstract
Translational control of mRNAs is a point of convergence for many oncogenic signals through which cancer cells tune protein expression in tumorigenesis. Cancer cells rely on translational control to appropriately adapt to limited resources while maintaining cell growth and survival, which creates a selective therapeutic window compared to non-transformed cells. In this review, we first discuss how cancer cells modulate the translational machinery to rapidly and selectively synthesize proteins in response to internal oncogenic demands and external factors in the tumor microenvironment. We highlight the clinical potential of compounds that target different translation factors as anti-cancer therapies. Next, we detail how RNA sequence and structural elements interface with the translational machinery and RNA-binding proteins to coordinate the translation of specific pro-survival and pro-growth programs. Finally, we provide an overview of the current and emerging technologies that can be used to illuminate the mechanisms of selective translational control in cancer cells as well as within the microenvironment.
Collapse
Affiliation(s)
- Joanna R Kovalski
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of UrologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Duygu Kuzuoglu‐Ozturk
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of UrologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of UrologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of Cellular and Molecular PharmacologyUniversity of California, San FranciscoSan FranciscoCAUSA
| |
Collapse
|
26
|
Ko S, German CA, Jensen A, Shen J, Wang A, Mehrotra DV, Sun YV, Sinsheimer JS, Zhou H, Zhou JJ. GWAS of longitudinal trajectories at biobank scale. Am J Hum Genet 2022; 109:433-445. [PMID: 35196515 PMCID: PMC8948167 DOI: 10.1016/j.ajhg.2022.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Biobanks linked to massive, longitudinal electronic health record (EHR) data make numerous new genetic research questions feasible. One among these is the study of biomarker trajectories. For example, high blood pressure measurements over visits strongly predict stroke onset, and consistently high fasting glucose and Hb1Ac levels define diabetes. Recent research reveals that not only the mean level of biomarker trajectories but also their fluctuations, or within-subject (WS) variability, are risk factors for many diseases. Glycemic variation, for instance, is recently considered an important clinical metric in diabetes management. It is crucial to identify the genetic factors that shift the mean or alter the WS variability of a biomarker trajectory. Compared to traditional cross-sectional studies, trajectory analysis utilizes more data points and captures a complete picture of the impact of time-varying factors, including medication history and lifestyle. Currently, there are no efficient tools for genome-wide association studies (GWASs) of biomarker trajectories at the biobank scale, even for just mean effects. We propose TrajGWAS, a linear mixed effect model-based method for testing genetic effects that shift the mean or alter the WS variability of a biomarker trajectory. It is scalable to biobank data with 100,000 to 1,000,000 individuals and many longitudinal measurements and robust to distributional assumptions. Simulation studies corroborate that TrajGWAS controls the type I error rate and is powerful. Analysis of eleven biomarkers measured longitudinally and extracted from UK Biobank primary care data for more than 150,000 participants with 1,800,000 observations reveals loci that significantly alter the mean or WS variability.
Collapse
Affiliation(s)
- Seyoon Ko
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher A German
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aubrey Jensen
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Anran Wang
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University, Atlanta, GA 30322, USA
| | - Janet S Sinsheimer
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hua Zhou
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jin J Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
27
|
Jafarzadeh A, Noori M, Sarrafzadeh S, Tamehri Zadeh SS, Nemati M, Chatrabnous N, Jafarzadeh S, Hamblin MR, Jafari Najaf Abadi MH, Mirzaei H. MicroRNA-383: A tumor suppressor miRNA in human cancer. Front Cell Dev Biol 2022; 10:955486. [PMID: 36313570 PMCID: PMC9608775 DOI: 10.3389/fcell.2022.955486] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/23/2022] [Indexed: 02/05/2023] Open
Abstract
Downregulated expression of anti-tumor miR-383 has been found in many kinds of cancer. MiR-383 family members can directly target the 3'-untranslated region (3'-UTR) of the mRNA of some pro-tumor genes to attenuate several cancer-related processes, including cell proliferation, invasion, migration, angiogenesis, immunosuppression, epithelial-mesenchymal transition, glycolysis, chemoresistance, and the development of cancer stem cells, whilst promoting apoptosis. Functionally, miR-383 operates as a tumor inhibitor miRNA in many types of cancer, including breast cancer, hepatocellular carcinoma, gastric cancer, pancreatic cancer, colorectal cancer, esophageal cancer, lung cancer, head and neck cancer, glioma, medulloblastoma, melanoma, prostate cancer, cervical cancer, oral squamous cell carcinoma, thyroid cancer, and B-cell lymphoma. Both pro-tumor and anti-tumor effects have been attributed to miR-383 in ovarian cancer. However, only the pro-tumor effects of miR-383 were reported in cholangiocarcinoma. The restoration of miR-383 expression could be considered a possible treatment for cancer. This review discusses the anti-tumor effects of miR-383 in human cancers, emphasizing their downstream target genes and potential treatment approaches.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- *Correspondence: Abdollah Jafarzadeh, ; Mohammad Hassan Jafari Najaf Abadi, ; Hamed Mirzaei,
| | - Majid Noori
- Golestan Hospital Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Sarrafzadeh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Maryam Nemati
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Chatrabnous
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Mohammad Hassan Jafari Najaf Abadi
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- *Correspondence: Abdollah Jafarzadeh, ; Mohammad Hassan Jafari Najaf Abadi, ; Hamed Mirzaei,
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- *Correspondence: Abdollah Jafarzadeh, ; Mohammad Hassan Jafari Najaf Abadi, ; Hamed Mirzaei,
| |
Collapse
|
28
|
Becker AE, Wu PK, Park JI. eIF5A-Independent Role of DHPS in p21 CIP1 and Cell Fate Regulation. Int J Mol Sci 2021; 22:13187. [PMID: 34947982 PMCID: PMC8707118 DOI: 10.3390/ijms222413187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Deoxyhypusine synthase (DHPS) catalyzes the first step of hypusination of the elongation translation factor 5A (eIF5A), and these two proteins have an exclusive enzyme-substrate relationship. Here we demonstrate that DHPS has a role independent of eIF5A hypusination in A375 and SK-MEL-28 human melanoma cells, in which the extracellular signal regulated kinase 1/2 (ERK1/2) pathway is deregulated. We found that RNA interference of DHPS induces G0/G1 cell cycle arrest in association with increased p21CIP1 expression in these cells whereas eIF5A knockdown induces cell death without increasing p21CIP1 expression. Interestingly, p21CIP1 knockdown switched DHPS knockdown-induced growth arrest to cell death in these cells, suggesting a specific relation between DHPS and p21CIP1 in determining cell fate. Surprisingly, ectopic expression of DHPS-K329R mutant that cannot hypusinate eIF5A abrogated DHPS knockdown-induced p21CIP1 expression in these cells, suggesting a non-canonical role of DHPS underlying the contrasting effects of DHPS and eIF5A knockdowns. We also show that DHPS knockdown induces p21CIP1 expression in these cells by increasing CDKN1A transcription through TP53 and SP1 in an ERK1/2-dependent manner. These data suggest that DHPS has a role independent of its ability to hypusinate eIF5A in cells, which appears to be important for regulating p21CIP1 expression and cell fate.
Collapse
Affiliation(s)
| | | | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (A.E.B.); (P.-K.W.)
| |
Collapse
|
29
|
Kachaev ZM, Ivashchenko SD, Kozlov EN, Lebedeva LA, Shidlovskii YV. Localization and Functional Roles of Components of the Translation Apparatus in the Eukaryotic Cell Nucleus. Cells 2021; 10:3239. [PMID: 34831461 PMCID: PMC8623629 DOI: 10.3390/cells10113239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/15/2022] Open
Abstract
Components of the translation apparatus, including ribosomal proteins, have been found in cell nuclei in various organisms. Components of the translation apparatus are involved in various nuclear processes, particularly those associated with genome integrity control and the nuclear stages of gene expression, such as transcription, mRNA processing, and mRNA export. Components of the translation apparatus control intranuclear trafficking; the nuclear import and export of RNA and proteins; and regulate the activity, stability, and functional recruitment of nuclear proteins. The nuclear translocation of these components is often involved in the cell response to stimulation and stress, in addition to playing critical roles in oncogenesis and viral infection. Many components of the translation apparatus are moonlighting proteins, involved in integral cell stress response and coupling of gene expression subprocesses. Thus, this phenomenon represents a significant interest for both basic and applied molecular biology. Here, we provide an overview of the current data regarding the molecular functions of translation factors and ribosomal proteins in the cell nucleus.
Collapse
Affiliation(s)
- Zaur M. Kachaev
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Z.M.K.); (S.D.I.); (E.N.K.); (L.A.L.)
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Sergey D. Ivashchenko
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Z.M.K.); (S.D.I.); (E.N.K.); (L.A.L.)
| | - Eugene N. Kozlov
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Z.M.K.); (S.D.I.); (E.N.K.); (L.A.L.)
| | - Lyubov A. Lebedeva
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Z.M.K.); (S.D.I.); (E.N.K.); (L.A.L.)
| | - Yulii V. Shidlovskii
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Z.M.K.); (S.D.I.); (E.N.K.); (L.A.L.)
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354340 Sochi, Russia
- Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| |
Collapse
|
30
|
Wasson JA, Harris G, Keppler-Ross S, Brock TJ, Dar AR, Butcher RA, Fischer SEJ, Kagias K, Clardy J, Zhang Y, Mango SE. Neuronal control of maternal provisioning in response to social cues. SCIENCE ADVANCES 2021; 7:7/34/eabf8782. [PMID: 34417172 PMCID: PMC8378817 DOI: 10.1126/sciadv.abf8782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/30/2021] [Indexed: 05/03/2023]
Abstract
Mothers contribute cytoplasmic components to their progeny in a process called maternal provisioning. Provisioning is influenced by the parental environment, but the molecular pathways that transmit environmental cues between generations are not well understood. Here, we show that, in Caenorhabditis elegans, social cues modulate maternal provisioning to regulate gene silencing in offspring. Intergenerational signal transmission depends on a pheromone-sensing neuron and neuronal FMRFamide (Phe-Met-Arg-Phe)-like peptides. Parental FMRFamide-like peptide signaling dampens oxidative stress resistance and promotes the deposition of mRNAs for translational components in progeny, which, in turn, reduces gene silencing. This study identifies a previously unknown pathway for intergenerational communication that links neuronal responses to maternal provisioning. We suggest that loss of social cues in the parental environment represents an adverse environment that stimulates stress responses across generations.
Collapse
Affiliation(s)
| | - Gareth Harris
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
- Department of Biology, California State University Channel Islands, Camarillo, CA, USA
| | | | | | - Abdul R Dar
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Sylvia E J Fischer
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Konstantinos Kagias
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Cambridge, MA, USA
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| | - Susan E Mango
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
31
|
Pálfi P, Bakacsy L, Kovács H, Szepesi Á. Hypusination, a Metabolic Posttranslational Modification of eIF5A in Plants during Development and Environmental Stress Responses. PLANTS 2021; 10:plants10071261. [PMID: 34206171 PMCID: PMC8309165 DOI: 10.3390/plants10071261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022]
Abstract
Hypusination is a unique posttranslational modification of eIF5A, a eukaryotic translation factor. Hypusine is a rare amino acid synthesized in this process and is mediated by two enzymes, deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH). Despite the essential participation of this conserved eIF5A protein in plant development and stress responses, our knowledge of its proper function is limited. In this review, we demonstrate the main findings regarding how eIF5A and hypusination could contribute to plant-specific responses in growth and stress-related processes. Our aim is to briefly discuss the plant-specific details of hypusination and decipher those signal pathways which can be effectively modified by this process. The diverse functions of eIF5A isoforms are also discussed in this review.
Collapse
|
32
|
Neelagandan N, Lamberti I, Carvalho HJF, Gobet C, Naef F. What determines eukaryotic translation elongation: recent molecular and quantitative analyses of protein synthesis. Open Biol 2020; 10:200292. [PMID: 33292102 PMCID: PMC7776565 DOI: 10.1098/rsob.200292] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
Protein synthesis from mRNA is an energy-intensive and tightly controlled cellular process. Translation elongation is a well-coordinated, multifactorial step in translation that undergoes dynamic regulation owing to cellular state and environmental determinants. Recent studies involving genome-wide approaches have uncovered some crucial aspects of translation elongation including the mRNA itself and the nascent polypeptide chain. Additionally, these studies have fuelled quantitative and mathematical modelling of translation elongation. In this review, we provide a comprehensive overview of the key determinants of translation elongation. We discuss consequences of ribosome stalling or collision, and how the cells regulate translation in case of such events. Next, we review theoretical approaches and widely used mathematical models that have become an essential ingredient to interpret complex molecular datasets and study translation dynamics quantitatively. Finally, we review recent advances in live-cell reporter and related analysis techniques, to monitor the translation dynamics of single cells and single-mRNA molecules in real time.
Collapse
Affiliation(s)
| | | | | | | | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| |
Collapse
|
33
|
Ning L, Wang L, Zhang H, Jiao X, Chen D. Eukaryotic translation initiation factor 5A in the pathogenesis of cancers. Oncol Lett 2020; 20:81. [PMID: 32863914 PMCID: PMC7436936 DOI: 10.3892/ol.2020.11942] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is the leading cause of death worldwide. The absence of obvious symptoms and insufficiently sensitive biomarkers in early stages of carcinoma limits early diagnosis. Cancer therapy agents and targeted therapy have been used extensively against tissues or organs of specific cancers. However, the intrinsic and/or acquired resistance to the agents or targeted drugs as well as the serious toxic side effects of the drugs would limit their use. Therefore, identifying biomarkers involved in tumorigenesis and progression represents a challenge for cancer diagnosis and therapeutic strategy development. The eukaryotic translation factor 5A (eIF5A), originally identified as an initiation factor, was later shown to promote translation elongation of iterated proline sequences. There are two eIF5A isoforms (eIF5A1 and eIF5A2). eIF5A2 protein consists of 153 residues, and shares 84% amino acid identity with eIF5A1. However, the biological functions of these two isoforms may be significantly different. Recently, it was demonstrated that eIF5Ais widely involved in the pathogenesis of a number of diseases, including cancers. In particular, eIF5A plays an important role in regulating tumor growth, invasion, metastasis and tumor microenvironment. It was also shown to serve as a potential biomarker and target for the diagnosis and treatment of cancers. The present review briefly discusses the latest findings of eIF5A in the pathogenesis of certain malignant cancers and evolving clinical applications.
Collapse
Affiliation(s)
- Liang Ning
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lei Wang
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Honglai Zhang
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xuelong Jiao
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Dong Chen
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|