1
|
Zhou W, Ruan H, Zhu L, Chen S, Yang M. Unveiling a Novel Glioblastoma Deep Molecular Profiling: Insight into the Cancer Cell Differentiation-Related Mechanisms. ACS OMEGA 2025; 10:10230-10250. [PMID: 40124014 PMCID: PMC11923693 DOI: 10.1021/acsomega.4c09586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND the sophisticated cellular heterogeneity of cell populations in glioblastoma (GBM) has been a key factor influencing tumor progression and response to therapy. The lack of more precise stratification based on cellular differentiation status poses a great challenge to therapeutic strategies. MATERIALS AND METHODS harnessing the bulk multiomics and single-nucleus RNA sequencing data available from the National Center for Biotechnology Information (NCBI) and The Cancer Genome Atlas (TCGA) Program repositories, we developed a novel and accurate GBM risk classification using an ensemble consensus clustering approach based on the junction of prognosis and trajectory analysis. Comprehensive cluster labeling and multiomics data characterization were also performed. RESULTS a novel GBM stratification model was constructed using 45 malignant cell fate genes: (a) energy metabolism-enhanced-type GBM; (b) invasion-enhanced-type GBM; (c) invasion-attenuated-type GBM; and (d) glycolysis-dominant energy metabolism-enhanced-type GBM. The biological plausibility of the model was verified through a range of comprehensive analyses of multiomics data, showing that cases with invasion-attenuated-type were the best prognosis and energy metabolism-enhanced-type the poorest. CONCLUSIONS the study has uncovered GBM complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying tumorigenesis. This precise stratification system provided implications for further studies of individual therapies.
Collapse
Affiliation(s)
- Weili Zhou
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Hongtao Ruan
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Lihua Zhu
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shunqiang Chen
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Muyi Yang
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| |
Collapse
|
2
|
Feng H, Yao W, Zhang Y, Liu Y, Liu B, Zhou J, Li J, Jiang Z, Ling F, Zhou J, Wu D, Li Y, Yang J, Zheng J. LGALS9B stabilizes EEF1D protein and activates the PI3K/AKT signaling pathway to promote gastric cancer occurrence and metastasis. Oncogene 2025; 44:652-664. [PMID: 39639171 DOI: 10.1038/s41388-024-03247-2] [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: 07/29/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Gastric cancer ranks among the most prevalent malignancies globally, characterized by limited treatment efficacy and high recurrence rates. Effective management of this disease requires a comprehensive understanding of its underlying pathogenic mechanisms. Galectins have emerged as promising targets in gastric cancer therapy, with Galectin-9 (LGALS9) receiving considerable attention in recent years. However, Galectin-9B (LGALS9B) remains relatively under-explored in gastric cancer research. Our study investigates the role of LGALS9B in gastric cancer progression, demonstrating that its over-expression enhances cellular proliferation, migration, and invasion, while its knockdown inhibits these processes both in vitro and in vivo. We further elucidate that LGALS9B competes with the E3 ligase HERC5 for binding to eukaryotic translation elongation factor 1 delta (EEF1D), thereby preventing its protein degradation. This interaction results in the enrichment of EEF1D, which activates the PI3K/AKT signaling pathway and ultimately promotes gastric cancer progression. These findings highlight the regulatory role of LGALS9B in the pathogenesis of gastric cancer, offering valuable insights into potential novel therapeutic strategies for managing this challenging disease.
Collapse
Affiliation(s)
- Huolun Feng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Wei Yao
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Yucheng Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yongfeng Liu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bin Liu
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Gannan Medical University, Ganzhou, China
| | - Ji Zhou
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiehui Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhuosheng Jiang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Fa Ling
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jianlong Zhou
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Deqing Wu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China.
| | - Juan Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| | - Jiabin Zheng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China.
| |
Collapse
|
3
|
Amiot J, Gubeljak L, Fontaine A, Smith D, Mortemousque I, Parodi N, Mauillon J, Kasper E, Baert-Desurmont S, Tinat J, Houdayer C. New RPS20 gene variant in colorectal cancer diagnosis: insight from a large series of patients. Fam Cancer 2025; 24:22. [PMID: 39920491 DOI: 10.1007/s10689-025-00446-y] [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: 11/28/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
Germline pathogenic variants of the RPS20 (ribosomal protein S20) gene are suspected to be involved in the predisposition to familial colorectal cancer (CRC) with no DNA mismatch repair deficiency. RPS20 pathogenic variants are very rare with only five reported cases in the literature. We report in this work the retrospective germline analysis of RPS20 for 1035 consecutive patients with a personal and/or familial history suggestive of hereditary predisposition to CRC. Within this series, a pathogenic variant in known CRC genes was found in 15% of cases and we describe one RPS20 loss-of-function variant (NM_001146227.1:c.115_116del, p.(Leu39Aspfs*33)). This frameshift is the first reported de novo variant in CRC, it was identified in in a female patient diagnosed with rectal cancer at the age of 35, 11 adenomatous polyps in 5 years and breast cancer at the age of 43. RPS20 has an intriguing role in oncogenesis, acting as an oncogene or tumour suppressor depending on the context, and is also involved in Diamond-Blackfan anemia via gain of function or dominant negative variants. This is therefore a complex gene for genetic counselling and, given the rarity of RPS20 pathogenic variants, we emphasise the need to collect data to clarify the phenotypic spectrum of RPS20-associated cancers and thus improve management.
Collapse
Affiliation(s)
- Julie Amiot
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, F-76000, France.
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France.
- Department of Genetics, Rouen Normandy University Hospital, Inserm U1245 Cancer and Brain Genomics, FHU- G4 Génomique UFR Santé, 22 Boulevard Gambetta, Rouen Cedex, 76183, France.
| | - Lara Gubeljak
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| | - Agathe Fontaine
- Department of Medical Genetics, CHU Bordeaux, Bordeaux, F-33000, France
| | - Denis Smith
- Department of Digestive Oncology, CHU Bordeaux, Bordeaux, F-33000, France
| | | | - Nathalie Parodi
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| | - Jacques Mauillon
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| | - Edwige Kasper
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, F-76000, France
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| | - Stéphanie Baert-Desurmont
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, F-76000, France
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| | - Julie Tinat
- Department of Medical Genetics, CHU Bordeaux, Bordeaux, F-33000, France
| | - Claude Houdayer
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Rouen, F-76000, France
- Department of Genetics, Univ Rouen Normandie, Normandie Univ, CHU Rouen, FHU-G4 Génomique, ERN GENTURIS, Rouen, F-76000, France
| |
Collapse
|
4
|
Thai A, Doescher C, Kamal N, Teramoto D, Fung C, Cha E, La V, Cheng P, Sedighim S, Keklikian A, Thankam FG. Single cell transcriptomics profiling of the stromal cells in the pathologic association of ribosomal proteins in the ischemic myocardium and epicardial fat. Cell Tissue Res 2025; 399:173-192. [PMID: 39641799 PMCID: PMC11787193 DOI: 10.1007/s00441-024-03933-3] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Sustenance of ischemia in the surviving cardiac tissue following myocardial infarction (MI) elicits a proinflammatory milieu resulting in subsequent pathological episodes. Also, the activation and release of ribosomal proteins under ischemic insults have been unveiled; however, their extra ribosomal functions are unknown. We identified the ribosomal proteins including RPL10A, RPL14, RPL30, RPS18, FAU-40 (RPS30), and RPSA (Laminin Receptor, LR) in the vesicles of ischemia challenged epicardial adipose tissue derived stromal cells (EATDS). The present study aimed to assess the association of these proteins in the epicardial adipose tissues (EAT) and left ventricular (LV) myocardium and isolated stromal cells (EATDS and LVSCs) from hyperlipidemic (HL), MI and coronary artery bypass graft (CABG) swine models. The findings revealed an upregulation of RPL10A, RPL14, RPL30, RPS18, RPS30, and RPSA in the LV tissues of CABG and HL swine with a concomitant reduction in the MI group. RPS30 displayed similar upregulation in EAT, whereas the expression of other ribosomal proteins was not significantly altered. Additionally, the ischemic LVSCs and EATDS displayed altered expression status of these genes compared to the control. Also, the RPS18 + , RPL30 + and RPSA + LVSCs favored ischemia and revealed similar anti-inflammatory and regenerative sub-phenotypes reflecting the protective/survival mechanisms. Further understanding regarding the underlying molecular mechanisms and functions of these ribosomal proteins offers immense translational opportunities in the better management of ischemic cardiac complications.
Collapse
Affiliation(s)
- An Thai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Christian Doescher
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Nawfal Kamal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Darren Teramoto
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Cameron Fung
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Ed Cha
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Vy La
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Pauline Cheng
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Sharona Sedighim
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Angelo Keklikian
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Finosh G Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
5
|
Wang R, Lv C, Li D, Song Y, Yan Z. EEF1D stabilized by SRSF9 promotes colorectal cancer via enhancing the proliferation and metastasis. Int J Cancer 2024; 155:1487-1499. [PMID: 38771720 DOI: 10.1002/ijc.35039] [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: 11/28/2023] [Revised: 03/21/2024] [Accepted: 04/16/2024] [Indexed: 05/23/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer and causes high mortality worldwide. Although CRC has been studied widely, the molecular mechanism is not completely known. Eukaryotic translation elongation factor 1 delta (EEF1D) participates in the progression of various tumors, however, the effect of EEF1D on CRC remains unclear. Here, we aimed to identify the potential mechanism of EEF1D in CRC. The expression levels of EEF1D were assessed in CRC samples. Functional analysis of EEF1D in CRC was detected in vitro and in vivo. The regulatory mechanism of EEF1D was identified with RNA immunoprecipitation, RNA pull-down assay, and proteomics analysis. Our findings confirmed that EEF1D was upregulated in human CRC tissues. Functionally, EEF1D overexpression accelerated cell proliferation and metastasis, whereas EEF1D knockdown inhibited cell proliferation and metastasis both in vitro and in vivo CRC models. Furthermore, we showed that EEF1D was upregulated by SRSF9 via binding to 3'UTR of EEF1D mRNA. EEF1D knockdown reversed the malignant phenotype induced by SRSF9 overexpression. These findings demonstrated that EEF1D promotes CRC progression, and EEF1D may be a molecular target against CRC.
Collapse
Affiliation(s)
- Rui Wang
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chi Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Donghao Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutong Song
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhaopeng Yan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa-SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [DOI: https:/doi.org/10.3390/biomedicines12020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3−3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
7
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa- SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [PMID: 38397997 PMCID: PMC11154313 DOI: 10.3390/biomedicines12020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 02/25/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3-3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico;
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico;
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
8
|
Guo L, Ren H, Fan S, Chao X, Liu M, Guan H, Wang J. Autoantibodies against eukaryotic translation elongation factor 1 delta in two patients with autoimmune cerebellar ataxia. Front Immunol 2024; 14:1289175. [PMID: 38332912 PMCID: PMC10850295 DOI: 10.3389/fimmu.2023.1289175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/26/2023] [Indexed: 02/10/2024] Open
Abstract
Background Autoantibodies are useful biomarkers for the early detection and diagnosis of autoimmune cerebellar ataxia (ACA). Objective To identify novel autoantibody candidates in ACA patients. Methods Patients with cerebellar ataxia of unknown cause were recruited from July 2018 to February 2023. Anti-neural autoantibodies in patient samples were detected by tissue-based indirect immunofluorescence assay (TBA) on rat cerebellum sections. TBA-positive samples were further screened for well-established anti-neural autoantibodies using commercial kits. Tissue-immunoprecipitation (TIP) and subsequent mass spectrometric (MS) analysis were used to explore the target antigens of autoantibodies in samples that were TBA-positive but negative for known autoantibodies. The specific binding between autoantibodies and the identified target antigen was confirmed by neutralization experiments, recombinant cell-based indirect immunofluorescence assay (CBA), and western blotting experiments. Results The eukaryotic translation elongation factor 1 delta (EEF1D) protein was identified as a target antigen of autoantibodies in samples from a 43-year-old female ACA patient, while the specific binding of autoantibodies and EEF1D was confirmed by subsequent experiments. A second anti-EEF1D autoantibody-positive ACA patient, a 59-year-old female, was detected in simultaneous screening. The main clinical manifestations in each of the two patients were cerebellar syndrome, such as unsteady walking and limb ataxia. Both patients received immunotherapy, including corticosteroids, intravenous immunoglobulin, and mycophenolate mofetil. Their outcomes provided evidence to support the effectiveness of immunotherapy, but the cerebellar atrophy that occurred before treatment may be irreversible. Conclusion In the current study, we identified anti-EEF1D autoantibody as a novel autoantibody candidate in ACA. Its pathological roles and diagnostic value need to be further verified in larger-scale studies.
Collapse
Affiliation(s)
- Liyuan Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Haitao Ren
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siyuan Fan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingchen Chao
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Mange Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongzhi Guan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Myers BL, Brayer KJ, Paez-Beltran LE, Keith MS, Suzuki H, Newville J, Anderson RH, Lo Y, Mertz CM, Kollipara R, Borromeo MD, Bachoo RM, Johnson JE, Vue TY. Glioblastoma initiation, migration, and cell types are regulated by core bHLH transcription factors ASCL1 and OLIG2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560206. [PMID: 37873200 PMCID: PMC10592871 DOI: 10.1101/2023.09.30.560206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Glioblastomas (GBMs) are highly aggressive, infiltrative, and heterogeneous brain tumors driven by complex driver mutations and glioma stem cells (GSCs). The neurodevelopmental transcription factors ASCL1 and OLIG2 are co-expressed in GBMs, but their role in regulating the heterogeneity and hierarchy of GBM tumor cells is unclear. Here, we show that oncogenic driver mutations lead to dysregulation of ASCL1 and OLIG2, which function redundantly to initiate brain tumor formation in a mouse model of GBM. Subsequently, the dynamic levels and reciprocal binding of ASCL1 and OLIG2 to each other and to downstream target genes then determine the cell types and degree of migration of tumor cells. Single-cell RNA sequencing (scRNA-seq) reveals that a high level of ASCL1 is key in defining GSCs by upregulating a collection of ribosomal protein, mitochondrial, neural stem cell (NSC), and cancer metastasis genes - all essential for sustaining the high proliferation, migration, and therapeutic resistance of GSCs.
Collapse
|
10
|
Wei A, Wang L. Prediction of Synaptically Localized RNAs in Human Neurons Using Developmental Brain Gene Expression Data. Genes (Basel) 2022; 13:1488. [PMID: 36011399 PMCID: PMC9408096 DOI: 10.3390/genes13081488] [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: 07/12/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
In the nervous system, synapses are special and pervasive structures between axonal and dendritic terminals, which facilitate electrical and chemical communications among neurons. Extensive studies have been conducted in mice and rats to explore the RNA pool at synapses and investigate RNA transport, local protein synthesis, and synaptic plasticity. However, owing to the experimental difficulties of studying human synaptic transcriptomes, the full pool of human synaptic RNAs remains largely unclear. We developed a new machine learning method, called PredSynRNA, to predict the synaptic localization of human RNAs. Training instances of dendritically localized RNAs were compiled from previous rodent studies, overcoming the shortage of empirical instances of human synaptic RNAs. Using RNA sequence and gene expression data as features, various models with different learning algorithms were constructed and evaluated. Strikingly, the models using the developmental brain gene expression features achieved superior performance for predicting synaptically localized RNAs. We examined the relevant expression features learned by PredSynRNA and used an independent test dataset to further validate the model performance. PredSynRNA models were then applied to the prediction and prioritization of candidate RNAs localized to human synapses, providing valuable targets for experimental investigations into neuronal mechanisms and brain disorders.
Collapse
Affiliation(s)
- Anqi Wei
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| | - Liangjiang Wang
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| |
Collapse
|
11
|
Tinterri C, Gentile D, Gatzemeier W, Sagona A, Barbieri E, Testori A, Errico V, Bottini A, Marrazzo E, Dani C, Dozin B, Boni L, Bruzzi P, Fernandes B, Franceschini D, Spoto R, Torrisi R, Scorsetti M, Santoro A, Canavese G. Preservation of Axillary Lymph Nodes Compared with Complete Dissection in T1-2 Breast Cancer Patients Presenting One or Two Metastatic Sentinel Lymph Nodes: The SINODAR-ONE Multicenter Randomized Clinical Trial. Ann Surg Oncol 2022; 29:5732-5744. [PMID: 35552930 DOI: 10.1245/s10434-022-11866-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/20/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The SINODAR-ONE trial is a prospective noninferiority multicenter randomized study aimed at assessing the role of axillary lymph node dissection (ALND) in patients undergoing either breast-conserving surgery or mastectomy for T1-2 breast cancer (BC) and presenting one or two macrometastatic sentinel lymph nodes (SLNs). The endpoints were to evaluate whether SLN biopsy (SLNB) only was associated with worsening of the prognosis compared with ALND in terms of overall survival (OS) and relapse. METHODS Patients were randomly assigned (1:1 ratio) to either removal of ≥ 10 axillary level I/II non-SLNs followed by adjuvant therapy (standard arm) or no further axillary treatment (experimental arm). RESULTS The trial started in April 2015 and ceased in April 2020, involving 889 patients. Median follow-up was 34.0 months. There were eight deaths (ALND, 4; SNLB only, 4), with 5-year cumulative mortality of 5.8% and 2.1% in the standard and experimental arm, respectively (p = 0.984). There were 26 recurrences (ALND 11; SNLB only, 15), with 5-year cumulative incidence of recurrence of 6.9% and 3.3% in the standard and experimental arm, respectively (p = 0.444). Only one axillary lymph node recurrence was observed in each arm. The 5-year OS rates were 98.9% and 98.8%, in the ALND and SNLB-only arm, respectively (p = 0.936). CONCLUSIONS The 3-year survival and relapse rates of T1-2 BC patients with one or two macrometastatic SLNs treated with SLNB only, and adjuvant therapy, were not inferior to those of patients treated with ALND. These results do not support the use of routine ALND.
Collapse
Affiliation(s)
- Corrado Tinterri
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Damiano Gentile
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | | | - Andrea Sagona
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Erika Barbieri
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alberto Testori
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valentina Errico
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alberto Bottini
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Carla Dani
- Department of Epidemiology, Biostatistics and Clinical Trials, IRCCS S. Martino, IST, Genoa, Italy
| | - Beatrice Dozin
- Department of Epidemiology, Biostatistics and Clinical Trials, IRCCS S. Martino, IST, Genoa, Italy
| | - Luca Boni
- Department of Epidemiology, Biostatistics and Clinical Trials, IRCCS S. Martino, IST, Genoa, Italy
| | - Paolo Bruzzi
- Department of Epidemiology, Biostatistics and Clinical Trials, IRCCS S. Martino, IST, Genoa, Italy
| | - Bethania Fernandes
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Davide Franceschini
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ruggero Spoto
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Rosalba Torrisi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marta Scorsetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppe Canavese
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | |
Collapse
|
12
|
Xu H, Yu S, Peng K, Gao L, Chen S, Shen Z, Han Z, Chen M, Lin J, Chen S, Kang M. The role of EEF1D in disease pathogenesis: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1600. [PMID: 34790806 PMCID: PMC8576685 DOI: 10.21037/atm-21-5025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/16/2021] [Indexed: 11/17/2022]
Abstract
Objective The purpose of this paper was to investigate the role and mechanism of EEF1D in various diseases, especially in tumorigenesis and development, and explore the possibility of EEF1D as a biological target. Background EEF1D is a part of the EEF1 protein complex, which can produce four protein isoforms, of which three short isoforms are used as translation elongation factors. The three short isoforms play a role in anti-aging, regulating the cell cycle, and promoting the occurrence and development of malignant tumors, and the only long-form isoform plays a role in the development of the nervous system. Methods We searched the PubMed and Web of Science databases for literature up to January 2021 using relevant keywords, including “EEF1D”, “eukaryotic translation elongation factor 1 delta”, “translation elongation factor”, “translation elongation factor and cancer”, and “translation elongation factor and nervous system disease”. We then created an overview of the literature and summarized the results of the paper. Conclusions Through the review of relevant articles, we found that EEF1D is obviously overexpressed in a variety of tumors, and can regulate the proliferation of tumor cells and tumor growth, as well as play a role in tumor invasion. EEF1D is likely to become a new biological target for tumor therapy and diagnosis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shaobin Yu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Kaiming Peng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lei Gao
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Sui Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhimin Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ziyang Han
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingduan Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jihong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shuchen Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Science, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Charwudzi A, Meng Y, Hu L, Ding C, Pu L, Li Q, Xu M, Zhai Z, Xiong S. Integrated bioinformatics analysis reveals dynamic candidate genes and signaling pathways involved in the progression and prognosis of diffuse large B-cell lymphoma. PeerJ 2021; 9:e12394. [PMID: 34760386 PMCID: PMC8570165 DOI: 10.7717/peerj.12394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
Background Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous malignancy with varied outcomes. However, the fundamental mechanisms remain to be fully defined. Aim We aimed to identify core differentially co-expressed hub genes and perturbed pathways relevant to the pathogenesis and prognosis of DLBCL. Methods We retrieved the raw gene expression profile and clinical information of GSE12453 from the Gene Expression Omnibus (GEO) database. We used integrated bioinformatics analysis to identify differentially co-expressed genes. The CIBERSORT analysis was also applied to predict tumor-infiltrating immune cells (TIICs) in the GSE12453 dataset. We performed survival and ssGSEA (single-sample Gene Set Enrichment Analysis) (for TIICs) analyses and validated the hub genes using GEPIA2 and an independent GSE31312 dataset. Results We identified 46 differentially co-expressed hub genes in the GSE12453 dataset. Gene expression levels and survival analysis found 15 differentially co-expressed core hub genes. The core genes prognostic values and expression levels were further validated in the GEPIA2 database and GSE31312 dataset to be reliable (p < 0.01). The core genes’ main KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichments were Ribosome and Coronavirus disease-COVID-19. High expressions of the 15 core hub genes had prognostic value in DLBCL. The core genes showed significant predictive accuracy in distinguishing DLBCL cases from non-tumor controls, with the area under the curve (AUC) ranging from 0.992 to 1.00. Finally, CIBERSORT analysis on GSE12453 revealed immune cells, including activated memory CD4+ T cells and M0, M1, and M2-macrophages as the infiltrates in the DLBCL microenvironment. Conclusion Our study found differentially co-expressed core hub genes and relevant pathways involved in ribosome and COVID-19 disease that may be potential targets for prognosis and novel therapeutic intervention in DLBCL.
Collapse
Affiliation(s)
- Alice Charwudzi
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ye Meng
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linhui Hu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chen Ding
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lianfang Pu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qian Li
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengling Xu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhimin Zhai
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shudao Xiong
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
14
|
Seo L, Kim YI, Kim H, Hyun K, Kim J, Lee JE. Discovery of Klf2 interactors in mouse embryonic stem cells by immunoprecipitation-mass spectrometry utilizing exogenously expressed bait. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140672. [PMID: 34000451 DOI: 10.1016/j.bbapap.2021.140672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/04/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
Krüppel-like factor 2 (Klf2) is a DNA-binding transcription factor that regulates embryonic stem cell-specific gene expression. Transcription cofactors such as p300 acetyltransferase and Erk kinases interact with Klf2, providing an additional layer of transcription regulation in embryonic stem cells. To carry out a thorough survey of the Klf2 interactome in embryonic stem cells and identify novel transcription cofactors, we designed a modified immunoprecipitation-mass spectrometry (IP-MS) method. In this method, recombinant Klf2, expressed and purified from Sf9 insect cells instead of ectopically expressed in cells, was used as bait. Using this modified IP-MS method, we discovered nine Klf2-interacting proteins, including the previously reported Crebbp and p300. These proteins showed at least an 8-fold increase in signal intensity in Klf2 pull-downs compared with controls, with P-values <0.010. Among the identified Klf2-binding proteins confirmed using our IP-MS workflow was Snd1, which we found to interact directly with Klf2 and function as a transcriptional coactivator of Klf2 to drive the Oct4 gene expression. Collectively, our IP-MS protocol may offer a useful tool for identifying novel transcription cofactors in stem cells.
Collapse
Affiliation(s)
- Lin Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Yong-In Kim
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon 34113, South Korea
| | - Hyoungmin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Kwangbeom Hyun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| | - J Eugene Lee
- Division of Policy and Strategy, Korea Research Institute of Standards and Science, Daejeon 34113, South Korea.
| |
Collapse
|
15
|
Hua X, Chen J, Ge S, Xiao H, Zhang L, Liang C. Integrated analysis of the functions of RNA binding proteins in clear cell renal cell carcinoma. Genomics 2020; 113:850-860. [PMID: 33169673 DOI: 10.1016/j.ygeno.2020.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/16/2020] [Indexed: 12/29/2022]
Abstract
RNA binding proteins (RBPs) dysregulation is involved in the processes of various tumors. However, the roles of RBPs in clear cell renal cell carcinoma (ccRCC) remain poorly understand. In present study, we first performed consensus clustering and identified two clusters, of which cluster 2 was closely correlated with the malignancy of ccRCC. Differentially expressed RBPs between normal and tumor tissues were obtained, comprising 71 up-regulated and 44 down-regulated ones. Then, ten hub genes were selected and validated using The Human Protein Atlas database and receiver operating characteristic curves, showing good diagnostic value for cancers. Besides, we identified ten RBPs with the most useful prognostic values, and were used to construct a risk score model. The model could be used to stratify patients with different prognosis and phenotype distributions. The model showed good performance and can be used as a complementation for clinical factors to guide clinical practice in the future.
Collapse
Affiliation(s)
- Xiaoliang Hua
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China; The Institute of Urology, Anhui Medical University, Hefei, China
| | - Juan Chen
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, 400016, Chongqing, China
| | - Shengdong Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China; The Institute of Urology, Anhui Medical University, Hefei, China
| | - Haibing Xiao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China; The Institute of Urology, Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China; The Institute of Urology, Anhui Medical University, Hefei, China.
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China; The Institute of Urology, Anhui Medical University, Hefei, China.
| |
Collapse
|
16
|
EEF1D Promotes Glioma Proliferation, Migration, and Invasion through EMT and PI3K/Akt Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7804706. [PMID: 33029523 PMCID: PMC7533006 DOI: 10.1155/2020/7804706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022]
Abstract
Eukaryotic translation elongation factor 1δ (EEF1D), a subunit of the elongation factor 1 complex of proteins, mediates the elongation process of protein synthesis. Besides this canonical role, EEF1D was found overexpressed in many tumors, like hepatocarcinomas and medulloblastomas. In the present study, we demonstrated for the first time that EEF1D may interact with other putative proteins to regulate cell proliferation, migration, and invasion through PI3K/Akt and EMT pathways in glioma. Furthermore, knockdown of EEF1D could reduce cell proliferation and impaired epithelial-mesenchymal transition (EMT) phenotypes, including cell invasion. Taken together, these results indicate that EEF1D and its partner proteins might play a critical role in glioma and serve as a potential therapeutic target of glioma.
Collapse
|
17
|
Domingueti CB, Castilho DAQ, de Oliveira CE, Janini JBM, González-Arriagada WA, Salo T, Coletta RD, Paranaíba LMR. Eukaryotic translation elongation factor 1δ, N-terminal propeptide of type I collagen and cancer-associated fibroblasts are prognostic markers of oral squamous cell carcinoma patients. Oral Surg Oral Med Oral Pathol Oral Radiol 2020; 130:700-707.e2. [PMID: 33132084 DOI: 10.1016/j.oooo.2020.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/06/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Identifying markers that influence oral squamous cell carcinoma (OSCC) prognosis is a fundamental strategy to improve the overall survival of patients. Markers such as eukaryotic translation elongation factor 1δ (EEF1D), fascin, N-terminal propeptide of type I collagen (PINP), and cancer-associated fibroblasts (CAFs) have been noticed in OSCCs and their levels are closely related to the prognosis of tumors. Our aim was to confirm the role of those markers in OSCC prognosis. STUDY DESIGN Immunohistochemistry was performed in 90 OSCC specimens. The associations between clinicopathologic features and expression of markers were assessed by χ2 test. Kaplan-Meier curves and univariate and multivariate Cox regression models were used for survival analysis. Markers were analyzed individually and in combination. RESULTS High expression of EEF1D (P = .017) and PINP (P = .02) and abundant density of CAFs in tumor stroma (P = .005) predicted significantly poor survival in OSCC patients. Multivariate analysis revealed that all 3 parameters are individually independent prognostic factors of OSCC patients, and their combination improved the discrimination of patients at high risk for poor survival. CONCLUSIONS Our results suggested that the expression of EEF1D and PINP and the density of CAFs might influence the survival of patients with OSCC.
Collapse
Affiliation(s)
- Catherine Bueno Domingueti
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Dayana Aparecida Queiroz Castilho
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Carine Ervolino de Oliveira
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | | | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Institute of Oral and Maxillofacial Disease, University of Helsinki, and HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | - Ricardo D Coletta
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Lívia Máris Ribeiro Paranaíba
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Wu D, Gao W, Li X, Tian C, Jiao N, Fang S, Xiao J, Xu Z, Zhu L, Zhang G, Zhu R. Dr AFC: drug repositioning through anti-fibrosis characteristic. Brief Bioinform 2020; 22:5860688. [PMID: 32572450 PMCID: PMC8138822 DOI: 10.1093/bib/bbaa115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a key component in the pathogenic mechanism of a variety of diseases. These diseases involving fibrosis may share common mechanisms and therapeutic targets, and therefore common intervention strategies and medicines may be applicable for these diseases. For this reason, deliberately introducing anti-fibrosis characteristics into predictive modeling may lead to more success in drug repositioning. In this study, anti-fibrosis knowledge base was first built by collecting data from multiple resources. Both structural and biological profiles were then derived from the knowledge base and used for constructing machine learning models including Structural Profile Prediction Model (SPPM) and Biological Profile Prediction Model (BPPM). Three external public data sets were employed for validation purpose and further exploration of potential repositioning drugs in wider chemical space. The resulting SPPM and BPPM models achieve area under the receiver operating characteristic curve (area under the curve) of 0.879 and 0.972 in the training set, and 0.814 and 0.874 in the testing set. Additionally, our results also demonstrate that substantial amount of multi-targeting natural products possess notable anti-fibrosis characteristics and might serve as encouraging candidates in fibrosis treatment and drug repositioning. To leverage our methodology and findings, we developed repositioning prediction platform, drug repositioning based on anti-fibrosis characteristic that is freely accessible via https://www.biosino.org/drafc.
Collapse
Affiliation(s)
| | | | | | - Chuan Tian
- Relay Therapeutics, Cambridge, United States
| | - Na Jiao
- Sun Yat-sen University, Guangzhou, China
| | - Sa Fang
- Tongji University, Shanghai, China
| | | | | | - Lixin Zhu
- Sun Yat-sen University, Guangzhou, China
| | - Guoqing Zhang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | | |
Collapse
|
19
|
Biterge-Sut B. Alterations in Eukaryotic Elongation Factor complex proteins (EEF1s) in cancer and their implications in epigenetic regulation. Life Sci 2019; 238:116977. [PMID: 31639400 DOI: 10.1016/j.lfs.2019.116977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/05/2019] [Accepted: 10/15/2019] [Indexed: 11/16/2022]
Abstract
AIMS In the cell, both transcriptional and translational processes are tightly regulated. Cancer is a multifactorial disease characterized by aberrant protein expression. Since epigenetic control mechanisms are also frequently disrupted during carcinogenesis, they have been the center of attention in cancer research within the past decades. EEF1 complex members, which are required for the elongation process in eukaryotes, have recently been implicated in carcinogenesis. This study aims to investigate genetic alterations within EEF1A1, EEF1A2, EEF1B2, EEF1D, EEF1E1 and EEF1G genes and their potential effects on epigenetic regulation mechanisms. MATERIALS AND METHODS In this study, we analyzed DNA sequencing and mRNA expression data available on The Cancer Genome Atlas (TCGA) across different cancer types to detect genetic alterations in EEF1 genes and investigated their potential impact on selected epigenetic modulators. KEY FINDINGS We found that EEF1 complex proteins were deregulated in several types of cancer. Lower EEF1A1, EEF1B2, EEF1D and EEF1G levels were correlated with poor survival in glioma, while lower EEF1B2, EEF1D and EEF1E1 levels were correlated with better survival in hepatocellular carcinoma. We detected genetic alterations within EEF1 genes in up to 35% of the patients and showed that these alterations resulted in down-regulation of histone modifying enzymes KMT2C, KMT2D, KMT2E, KAT6A and EP300. SIGNIFICANCE Here in this study, we showed that EEF1 deregulations might result in differential epigenomic landscapes, which affect the overall transcriptional profile, contributing to carcinogenesis. Identification of these molecular distinctions might be useful in developing targeted drug therapies and personalized medicine.
Collapse
Affiliation(s)
- Burcu Biterge-Sut
- Nigde Omer Halisdemir University, Faculty of Medicine, Department of Medical Biology, Nigde, Turkey.
| |
Collapse
|
20
|
Translation elongation factor eEF1Bα is identified as a novel prognostic marker of gastric cancer. Int J Biol Macromol 2018; 126:345-351. [PMID: 30572058 DOI: 10.1016/j.ijbiomac.2018.12.126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/15/2018] [Accepted: 12/15/2018] [Indexed: 11/24/2022]
Abstract
Gastric cancer (GC) is a common cancer in humans. Although overexpression of eukaryotic translation elongation factor eEF1Bα is associated with cancer onset and progression, little is known about its expression in GC and its prognostic significance. Here we used immunohistochemistry to analyze eEF1Bα expression in the following tissue types: GC, normal gastric, chronic gastritis, intestinal metaplasia, and intraepithelial neoplasia. These data were correlated with patients' clinical information. eEF1Bα was expressed at levels approximately three times higher in GC tissues compared with normal gastric tissues. High expression of eEF1Bα was significantly associated with histological type, TNM stage, tumor size, and distant metastases. GC patients with high eEF1Bα expression experienced significantly shorter overall survival. Bioinformatics analysis indicated that eEF1Bα may be associated with protein synthesis, energy metabolism, cell cycle, and the p53 signaling pathway. We identified the products of RPL10A and RPS13 as critical components of a network comprising eEF1Bα. We concluded that high eEF1Bα expression is associated with poor overall survival and may serve as an independent prognostic factor of GC. Further, we proposed that eEF1Bα likely mediates the development of GC through the cell cycle and p53 signaling pathway. Together, our findings suggest that eEF1Bα could be an effective prognostic biomarker for GC.
Collapse
|
21
|
Investigating Pathogenic and Hepatocarcinogenic Mechanisms from Normal Liver to HCC by Constructing Genetic and Epigenetic Networks via Big Genetic and Epigenetic Data Mining and Genome-Wide NGS Data Identification. DISEASE MARKERS 2018; 2018:8635329. [PMID: 30344796 PMCID: PMC6174771 DOI: 10.1155/2018/8635329] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/02/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022]
Abstract
The prevalence of hepatocellular carcinoma (HCC) is still high worldwide because liver diseases could develop into HCC. Recent reports indicate nonalcoholic fatty liver disease and nonalcoholic steatohepatitis (NAFLD&NASH) and primary biliary cirrhosis and primary sclerosing cholangitis (PBC&PSC) are significant of HCC. Therefore, understanding the cellular mechanisms of the pathogenesis and hepatocarcinogenesis from normal liver cells to HCC through NAFLD&NASH or PBC&PSC is a priority to prevent the progression of liver damage and reduce the risk of further complications. By the genetic and epigenetic data mining and the system identification through next-generation sequencing data and its corresponding DNA methylation profiles of liver cells in normal, NAFLD&NASH, PBC&PSC, and HCC patients, we identified the genome-wide real genetic and epigenetic networks (GENs) of normal, NAFLD&NASH, PBC&PSC, and HCC patients. In order to get valuable insight into these identified genome-wide GENs, we then applied a principal network projection method to extract the corresponding core GENs for normal liver cells, NAFLD&NASH, PBC&PSC, and HCC. By comparing the signal transduction pathways involved in the identified core GENs, we found that the hepatocarcinogenesis through NAFLD&NASH was induced through DNA methylation of HIST2H2BE, HSPB1, RPL30, and ALDOB and the regulation of miR-21 and miR-122, and the hepatocarcinogenesis through PBC&PSC was induced through DNA methylation of RPL23A, HIST2H2BE, TIMP1, IGF2, RPL30, and ALDOB and the regulation of miR-29a, miR-21, and miR-122. The genetic and epigenetic changes in the pathogenesis and hepatocarcinogenesis potentially serve as potential diagnostic biomarkers and/or therapeutic targets.
Collapse
|
22
|
Interplay between human nucleolar GNL1 and RPS20 is critical to modulate cell proliferation. Sci Rep 2018; 8:11421. [PMID: 30061673 PMCID: PMC6065441 DOI: 10.1038/s41598-018-29802-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/13/2018] [Indexed: 12/29/2022] Open
Abstract
Human Guanine nucleotide binding protein like 1 (GNL1) belongs to HSR1_MMR1 subfamily of nucleolar GTPases. Here, we report for the first time that GNL1 promotes cell cycle and proliferation by inducing hyperphosphorylation of retinoblastoma protein. Using yeast two-hybrid screening, Ribosomal protein S20 (RPS20) was identified as a functional interacting partner of GNL1. Results from GST pull-down and co-immunoprecipitation assays confirmed that interaction between GNL1 and RPS20 was specific. Further, GNL1 induced cell proliferation was altered upon knockdown of RPS20 suggesting its critical role in GNL1 function. Interestingly, cell proliferation was significantly impaired upon expression of RPS20 interaction deficient GNL1 mutant suggest that GNL1 interaction with RPS20 is critical for cell growth. Finally, the inverse correlation of GNL1 and RPS20 expression in primary colon and gastric cancers with patient survival strengthen their critical importance during tumorigenesis. Collectively, our data provided evidence that cross-talk between GNL1 and RPS20 is critical to promote cell proliferation.
Collapse
|
23
|
Bram Ednersson S, Stenson M, Stern M, Enblad G, Fagman H, Nilsson-Ehle H, Hasselblom S, Andersson PO. Expression of ribosomal and actin network proteins and immunochemotherapy resistance in diffuse large B cell lymphoma patients. Br J Haematol 2018; 181:770-781. [PMID: 29767447 DOI: 10.1111/bjh.15259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/05/2018] [Indexed: 10/25/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) patients with early relapse or refractory disease have a very poor outcome. Immunochemotherapy resistance will probably, also in the era of targeted drugs, remain the major cause of treatment failure. We used proteomic mass spectrometry to analyse the global protein expression of micro-dissected formalin-fixed paraffin-embedded tumour tissues from 97 DLBCL patients: 44 with primary refractory disease or relapse within 1 year from diagnosis (REF/REL), and 53 who were progression-free more than 5 years after diagnosis (CURED). We identified 2127 proteins: 442 were found in all patients and 102 were differentially expressed. Sixty-five proteins were overexpressed in REF/REL patients, of which 46 were ribosomal proteins (RPs) compared with 2 of the 37 overexpressed proteins in CURED patients (P = 7·6 × 10-10 ). Twenty of 37 overexpressed proteins in CURED patients were associated with actin regulation, compared with 1 of 65 in REF/REL patients (P = 1·4 × 10-9 ). Immunohistochemical staining showed higher expression of RPS5 and RPL17 in REF/REL patients while MARCKS-like protein, belonging to the actin network, was more highly expressed in CURED patients. Even though functional studies aimed at individual proteins and protein interactions to evaluate potential clinical effect are needed, our findings suggest new mechanisms behind immunochemotherapy resistance in DLBCL.
Collapse
Affiliation(s)
- Susanne Bram Ednersson
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Martin Stenson
- Section of Haematology, Department of Medicine, Kungälvs Hospital, Kungälv, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mimmie Stern
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology/Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Henrik Fagman
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Herman Nilsson-Ehle
- Section of Haematology and Coagulation, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sverker Hasselblom
- Department of Research, Development and Education, Region Halland, Halmstad, Sweden
| | - Per-Ola Andersson
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Flentje A, Kober KM, Carrico AW, Neilands TB, Flowers E, Heck NC, Aouizerat BE. Minority stress and leukocyte gene expression in sexual minority men living with treated HIV infection. Brain Behav Immun 2018; 70:335-345. [PMID: 29548994 PMCID: PMC5953835 DOI: 10.1016/j.bbi.2018.03.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/20/2018] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Sexual minority (i.e., non-heterosexual) individuals experience poorer mental and physical health, accounted for in part by the additional burden of sexual minority stress occurring from being situated in a culture favoring heteronormativity. Informed by previous research, the purpose of this study was to identify the relationship between sexual minority stress and leukocyte gene expression related to inflammation, cancer, immune function, and cardiovascular function. Sexual minority men living with HIV who were on anti-retroviral medication, had viral load < 200 copies/mL, and had biologically confirmed, recent methamphetamine use completed minority stress measures and submitted blood samples for RNA sequencing on leukocytes. Differential gene expression and pathway analyses were conducted comparing those with clinically elevated minority stress (n = 18) and those who did not meet the clinical cutoff (n = 20), covarying reactive urine toxicology results for very recent stimulant use. In total, 90 differentially expressed genes and 138 gene set pathways evidencing 2-directional perturbation were observed at false discovery rate (FDR) < 0.10. Of these, 41 of the differentially expressed genes and 35 of the 2-directionally perturbed pathways were identified as functionally related to hypothesized mechanisms of inflammation, cancer, immune function, and cardiovascular function. The neuroactive-ligand receptor pathway (implicated in cancer development) was identified using signaling pathway impact analysis. Our results suggest several potential biological pathways for future work investigating the relationship between sexual minority stress and health.
Collapse
Affiliation(s)
- Annesa Flentje
- Community Health Systems, School of Nursing, University of California, San Francisco, United States.
| | - Kord M Kober
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, United States; Institute for Computational Health Sciences, University of California, San Francisco, United States
| | | | - Torsten B Neilands
- Center for AIDS Prevention Studies, Department of Medicine, University of California, San Francisco, United States
| | - Elena Flowers
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, United States; Institute for Human Genetics, University of California, San Francisco, United States
| | - Nicholas C Heck
- Department of Psychology, Marquette University, United States
| | - Bradley E Aouizerat
- Bluestone Center for Clinical Research, College of Dentistry, New York University, United States
| |
Collapse
|
25
|
Cheng DD, Li SJ, Zhu B, Zhou SM, Yang QC. EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:50. [PMID: 29510727 PMCID: PMC5839064 DOI: 10.1186/s13046-018-0715-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/21/2018] [Indexed: 11/17/2022]
Abstract
Background Dysregulation of eukaryotic translation elongation factor 1 delta (EEF1D) in cancers has been reported; however, the role and mechanisms of EEF1D in osteosarcoma remain poorly understood. The aim of this study is to investigate the expression and role of EEF1D in osteosarcoma and to elucidate its underlying mechanisms. Methods The expression of EEF1D in osteosarcomas and cell lines was evaluated by qRT-PCR, Western blotting and immunohistochemistry. EEF1D knockdown using small interfering RNA (siRNA) was employed to analyze the role of EEF1D in osteosarcoma cell proliferation and cell cycle progression. The host signaling pathways affected by EEF1D knockdown were detected using PathScan® intracellular signaling array kit. Results The expression of EEF1D was found to be up-regulated in human osteosarcoma tissues and cell lines. Its expression was positively correlated with Enneking stage and the tumor recurrence. EEF1D knockdown inhibited osteosarcoma cell proliferation, colony-forming ability, and cell cycle G2/M transition in vitro. In addition, EEF1D knockdown decreased the levels of phospho-Akt, phospho-mTOR, and phospho-Bad proteins. Conclusions EEF1D is upregulated in osteosarcoma and plays a tumor promoting role by facilitating Akt-mTOR and Akt-Bad signaling pathways. Accordingly, EEF1D is a potential target for cancer therapy. Electronic supplementary material The online version of this article (10.1186/s13046-018-0715-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dong-Dong Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, China
| | - Shi-Jie Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, China
| | - Bin Zhu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, China
| | - Shu-Min Zhou
- Institution of microsurgery for limbs, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, China.
| | - Qing-Cheng Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
26
|
Hassan MK, Kumar D, Naik M, Dixit M. The expression profile and prognostic significance of eukaryotic translation elongation factors in different cancers. PLoS One 2018; 13:e0191377. [PMID: 29342219 PMCID: PMC5771626 DOI: 10.1371/journal.pone.0191377] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
Eukaryotic translation factors, especially initiation factors have garnered much attention with regards to their role in the onset and progression of different cancers. However, the expression levels and prognostic significance of translation elongation factors remain poorly explored in different cancers. In this study, we have investigated the mRNA transcript levels of seven translation elongation factors in different cancer types using Oncomine and TCGA databases. Furthermore, we have identified the prognostic significance of these factors using Kaplan-Meier Plotter and SurvExpress databases. We observed altered expression levels of all the elongation factors in different cancers. Higher expression of EEF1A2, EEF1B2, EEF1G, EEF1D, EEF1E1 and EEF2 was observed in most of the cancer types, whereas reverse trend was observed for EEF1A1. Overexpression of many factors predicted poor prognosis in breast (EEF1D, EEF1E1, EEF2) and lung cancer (EEF1A2, EEF1B2, EEF1G, EEF1E1). However, we didn’t see any common correlation of expression levels of elongation factors with survival outcomes across cancer types. Cancer subtype stratification showed association of survival outcomes and expression levels of elongation factors in specific sub-types of breast, lung and gastric cancer. Most interestingly, we observed a reciprocal relationship between the expression levels of the two EEF1A isoforms viz. EEF1A1 and EEF1A2, in most of the cancer types. Our results suggest that translation elongation factors can have a role in tumorigenesis and affect survival in cancer specific manner. Elongation factors have potential to serve as biomarkers and therapeutic drug targets, yet further study is required. Reciprocal relationship of differential expression between EEF1A isoforms observed in multiple cancer types indicates opposing roles in cancer and needs further investigation.
Collapse
Affiliation(s)
- Md. Khurshidul Hassan
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Bhimpur- Padanpur, Jatni, Khurda, Odisha, India
| | - Dinesh Kumar
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Bhimpur- Padanpur, Jatni, Khurda, Odisha, India
| | - Monali Naik
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Bhimpur- Padanpur, Jatni, Khurda, Odisha, India
| | - Manjusha Dixit
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Bhimpur- Padanpur, Jatni, Khurda, Odisha, India
- * E-mail:
| |
Collapse
|
27
|
Zhang YH, Huang T, Chen L, Xu Y, Hu Y, Hu LD, Cai Y, Kong X. Identifying and analyzing different cancer subtypes using RNA-seq data of blood platelets. Oncotarget 2017; 8:87494-87511. [PMID: 29152097 PMCID: PMC5675649 DOI: 10.18632/oncotarget.20903] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022] Open
Abstract
Detection and diagnosis of cancer are especially important for early prevention and effective treatments. Traditional methods of cancer detection are usually time-consuming and expensive. Liquid biopsy, a newly proposed noninvasive detection approach, can promote the accuracy and decrease the cost of detection according to a personalized expression profile. However, few studies have been performed to analyze this type of data, which can promote more effective methods for detection of different cancer subtypes. In this study, we applied some reliable machine learning algorithms to analyze data retrieved from patients who had one of six cancer subtypes (breast cancer, colorectal cancer, glioblastoma, hepatobiliary cancer, lung cancer and pancreatic cancer) as well as healthy persons. Quantitative gene expression profiles were used to encode each sample. Then, they were analyzed by the maximum relevance minimum redundancy method. Two feature lists were obtained in which genes were ranked rigorously. The incremental feature selection method was applied to the mRMR feature list to extract the optimal feature subset, which can be used in the support vector machine algorithm to determine the best performance for the detection of cancer subtypes and healthy controls. The ten-fold cross-validation for the constructed optimal classification model yielded an overall accuracy of 0.751. On the other hand, we extracted the top eighteen features (genes), including TTN, RHOH, RPS20, TRBC2, in another feature list, the MaxRel feature list, and performed a detailed analysis of them. The results indicated that these genes could be important biomarkers for discriminating different cancer subtypes and healthy controls.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China.,Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yu Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lan-Dian Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yudong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| |
Collapse
|
28
|
Lv H, Dong W, Qian G, Wang J, Li X, Cao Z, Lv Q, Wang C, Guo K, Zhang Y. uS10, a novel Npro-interacting protein, inhibits classical swine fever virus replication. J Gen Virol 2017; 98:1679-1692. [PMID: 28721853 DOI: 10.1099/jgv.0.000867] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Classical swine fever (CSF) is a severe, febrile and highly contagious disease caused by classical swine fever virus (CSFV) that has resulted in huge economic losses in the pig industry worldwide. CSFV Npro has been actively studied but remains incompletely understood. Few studies have investigated the cellular proteins that interact with Npro and their participation in viral replication. Here, the yeast two-hybrid (Y2H) system was employed to screen Npro-interacting proteins from a porcine alveolar macrophage (PAM) cDNA library, and a blast search of the NCBI database revealed that 15 cellular proteins interact with Npro. The interaction of Npro with ribosomal protein S20, also known as universal S10 (uS10), was further confirmed by co-immunoprecipitation and glutathione S-transferase pull-down assays. Furthermore, uS10 overexpression inhibited CSFV replication, whereas the knockdown of uS10 promoted CSFV replication in PAMs. In addition, Npro or CSFV reduced uS10 expression in PAMs in a proteasome-dependent manner, indicating that Npro-uS10 interaction might contribute to persistent CSFV replication. Our previous research showed that CSFV decreases Toll-like receptor 3 (TLR3) expression. The results showed that uS10 knockdown reduced TLR3 expression, and that uS10 overexpression increased TLR3 expression. Notably, uS10 knockdown did not promote CSFV replication following TLR3 overexpression. Conversely, uS10 overexpression did not inhibit CSFV replication following TLR3 knockdown. These results revealed that uS10 inhibits CSFV replication by modulating TLR3 expression. This work addresses a novel aspect of the regulation of the innate antiviral immune response during CSFV infection.
Collapse
Affiliation(s)
- Huifang Lv
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Wang Dong
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Gui Qian
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Jie Wang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Xiaomeng Li
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Zhi Cao
- Qingdao Yebio Biological Engineering Co. Ltd. (Yebio), No. 21 Aodongnan Road, Qingdao 266114, Shandong, PR China
| | - Qizhuang Lv
- College of Biology and Pharmacy, Yulin Normal University, No. 1303 Jiaoyu East Road, Yulin 537000, Guangxi, PR China
| | - Chengbao Wang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, No. 22 Xinong Road, Yangling 712100, Shaanxi, PR China
| |
Collapse
|
29
|
EEF1D modulates proliferation and epithelial-mesenchymal transition in oral squamous cell carcinoma. Clin Sci (Lond) 2016; 130:785-99. [PMID: 26823560 DOI: 10.1042/cs20150646] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
EEF1D (eukaryotic translation elongation factor 1δ) is a subunit of the elongation factor 1 complex of proteins that mediates the elongation process during protein synthesis via enzymatic delivery of aminoacyl-tRNAs to the ribosome. Although the functions of EEF1D in the translation process are recognized, EEF1D expression was found to be unbalanced in tumours. In the present study, we demonstrate the overexpression of EEF1D in OSCC (oral squamous cell carcinoma), and revealed that EEF1D and protein interaction partners promote the activation of cyclin D1 and vimentin proteins. EEF1D knockdown in OSCC reduced cell proliferation and induced EMT (epithelial-mesenchymal transition) phenotypes, including cell invasion. Taken together, these results define EEF1D as a critical inducer of OSCC proliferation and EMT.
Collapse
|
30
|
Silva MP, Barros-Silva JD, Vieira J, Lisboa S, Torres L, Correia C, Vieira-Coimbra M, Martins AT, Jerónimo C, Henrique R, Paulo P, Teixeira MR. NCOA2 is a candidate target gene of 8q gain associated with clinically aggressive prostate cancer. Genes Chromosomes Cancer 2016; 55:365-74. [PMID: 26799514 DOI: 10.1002/gcc.22340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/16/2015] [Accepted: 11/30/2015] [Indexed: 12/28/2022] Open
Abstract
Prostate carcinomas harboring 8q gains are associated with poor clinical outcome, but the target genes of this genomic alteration remain to be unveiled. In this study, we aimed to identify potential 8q target genes associated with clinically aggressive prostate cancer (PCa) using fluorescence in situ hybridization (FISH), genome-wide mRNA expression, and protein expression analyses. Using FISH, we first characterized the relative copy number of 8q (assessed with MYC flanking probes) of a series of 50 radical prostatectomy specimens, with available global gene expression data and typed for E26 transformation specific (ETS) rearrangements, and then compared the gene expression profile of PCa subsets with and without 8q24 gain using Significance Analysis of Microarrays. In the subset of tumors with ERG fusion genes (ERG+), five genes were identified as significantly overexpressed (false discovery rate [FDR], ≤ 5%) in tumors with relative 8q24 gain, namely VN1R1, ZNF417, CDON, IKZF2, and NCOA2. Of these, only NCOA2 is located in 8q (8q13.3), showing a statistically higher mRNA expression in the subgroup with relative 8q gain, both in the ERG+ subgroup and in the whole series (P = 0.000152 and P = 0.008, respectively). Combining all the cases with NCOA2 overexpression, either at the mRNA or at the protein level, we identified a group of tumors with NCOA2 copy-number increase, independently of ETS status and relative 8q24 gain. Furthermore, for the first time, we detected a structural rearrangement involving NCOA2 in PCa. These findings warrant further studies with larger series to evaluate if NCOA2 relative copy-number gain presents prognostic value independently of the well-established poor prognosis associated with MYC relative copy-number gain.
Collapse
Affiliation(s)
- Maria P Silva
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - João D Barros-Silva
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Joana Vieira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Susana Lisboa
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Lurdes Torres
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Cecília Correia
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Márcia Vieira-Coimbra
- Cancer Biology and Epigenetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Ana T Martins
- Cancer Biology and Epigenetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Paula Paulo
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
31
|
Yong WH, Shabihkhani M, Telesca D, Yang S, Tso JL, Menjivar JC, Wei B, Lucey GM, Mareninov S, Chen Z, Liau LM, Lai A, Nelson SF, Cloughesy TF, Tso CL. Ribosomal Proteins RPS11 and RPS20, Two Stress-Response Markers of Glioblastoma Stem Cells, Are Novel Predictors of Poor Prognosis in Glioblastoma Patients. PLoS One 2015; 10:e0141334. [PMID: 26506620 PMCID: PMC4624638 DOI: 10.1371/journal.pone.0141334] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/06/2015] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma stem cells (GSC) co-exhibiting a tumor-initiating capacity and a radio-chemoresistant phenotype, are a compelling cell model for explaining tumor recurrence. We have previously characterized patient-derived, treatment-resistant GSC clones (TRGC) that survived radiochemotherapy. Compared to glucose-dependent, treatment-sensitive GSC clones (TSGC), TRGC exhibited reduced glucose dependence that favor the fatty acid oxidation pathway as their energy source. Using comparative genome-wide transcriptome analysis, a series of defense signatures associated with TRGC survival were identified and verified by siRNA-based gene knockdown experiments that led to loss of cell integrity. In this study, we investigate the prognostic value of defense signatures in glioblastoma (GBM) patients using gene expression analysis with Probeset Analyzer (131 GBM) and The Cancer Genome Atlas (TCGA) data, and protein expression with a tissue microarray (50 GBM), yielding the first TRGC-derived prognostic biomarkers for GBM patients. Ribosomal protein S11 (RPS11), RPS20, individually and together, consistently predicted poor survival of newly diagnosed primary GBM tumors when overexpressed at the RNA or protein level [RPS11: Hazard Ratio (HR) = 11.5, p<0.001; RPS20: HR = 4.5, p = 0.03; RPS11+RPS20: HR = 17.99, p = 0.001]. The prognostic significance of RPS11 and RPS20 was further supported by whole tissue section RPS11 immunostaining (27 GBM; HR = 4.05, p = 0.01) and TCGA gene expression data (578 primary GBM; RPS11: HR = 1.19, p = 0.06; RPS20: HR = 1.25, p = 0.02; RPS11+RPS20: HR = 1.43, p = 0.01). Moreover, tumors that exhibited unmethylated O-6-methylguanine-DNA methyltransferase (MGMT) or wild-type isocitrate dehydrogenase 1 (IDH1) were associated with higher RPS11 expression levels [corr (IDH1, RPS11) = 0.64, p = 0.03); [corr (MGMT, RPS11) = 0.52, p = 0.04]. These data indicate that increased expression of RPS11 and RPS20 predicts shorter patient survival. The study also suggests that TRGC are clinically relevant cells that represent resistant tumorigenic clones from patient tumors and that their properties, at least in part, are reflected in poor-prognosis GBM. The screening of TRGC signatures may represent a novel alternative strategy for identifying new prognostic biomarkers.
Collapse
Affiliation(s)
- William H. Yong
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Maryam Shabihkhani
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Donatello Telesca
- Department of Biostatistics, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Shuai Yang
- Department of Neurosurgery, General Hospital of Guangzhou Military Command, Guangzhou, China
- Department of Surgery/Surgical-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jonathan L. Tso
- Department of Surgery/Surgical-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jimmy C. Menjivar
- Department of Surgery/Surgical-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Bowen Wei
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Gregory M. Lucey
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Sergey Mareninov
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Zugen Chen
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Linda M. Liau
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert Lai
- Department of Neurology/Neuro-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Stanley F. Nelson
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Timothy F. Cloughesy
- Department of Neurology/Neuro-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Cho-Lea Tso
- Department of Surgery/Surgical-Oncology, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Slavov N, Semrau S, Airoldi E, Budnik B, van Oudenaarden A. Differential Stoichiometry among Core Ribosomal Proteins. Cell Rep 2015; 13:865-73. [PMID: 26565899 PMCID: PMC4644233 DOI: 10.1016/j.celrep.2015.09.056] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/31/2015] [Accepted: 09/18/2015] [Indexed: 01/04/2023] Open
Abstract
Understanding the regulation and structure of ribosomes is essential to understanding protein synthesis and its dysregulation in disease. While ribosomes are believed to have a fixed stoichiometry among their core ribosomal proteins (RPs), some experiments suggest a more variable composition. Testing such variability requires direct and precise quantification of RPs. We used mass spectrometry to directly quantify RPs across monosomes and polysomes of mouse embryonic stem cells (ESC) and budding yeast. Our data show that the stoichiometry among core RPs in wild-type yeast cells and ESC depends both on the growth conditions and on the number of ribosomes bound per mRNA. Furthermore, we find that the fitness of cells with a deleted RP-gene is inversely proportional to the enrichment of the corresponding RP in polysomes. Together, our findings support the existence of ribosomes with distinct protein composition and physiological function. Wild-type yeast and mouse cells build ribosomes with different protein composition The stoichiometry among ribosomal proteins (RP) correlates to growth rate RP stoichiometry depends on the number of ribosomes bound per mRNA RP stoichiometry depends on the growth conditions
Collapse
Affiliation(s)
- Nikolai Slavov
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA; Department of Statistics and FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Stefan Semrau
- Leiden Institute of Physics, Leiden University, 2333 CC Leiden, the Netherlands
| | - Edoardo Airoldi
- Department of Statistics and FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Bogdan Budnik
- Department of Statistics and FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| |
Collapse
|
33
|
Liu K, Ren Y, Pang L, Qi Y, Jia W, Tao L, Hu Z, Zhao J, Zhang H, Li L, Yue H, Han J, Liang W, Hu J, Zou H, Yuan X, Li F. Papillary renal cell carcinoma: a clinicopathological and whole-genome exon sequencing study. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:8311-8335. [PMID: 26339402 PMCID: PMC4555730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/26/2015] [Indexed: 06/05/2023]
Abstract
Papillary renal cell carcinoma (PRCC) represents the second most common histological subtype of RCC, and comprises 2 subtypes. Prognosis for type 1 PRCC is relatively good, whereas type 2 PRCC is associated with poor clinical outcomes. The aim of the present study was to evaluate the clinicopathological and mutations characteristics of PRCC. Hence, we reported on 13 cases of PRCC analyzed using whole-exome sequencing. Histologically, type 2 PRCC showed a higher nuclear grade and lymphovascular invasion rate versus type 1 PRCC (P < 0.05). Immunostaining revealed type 1 PRCC had higher CK7 and lower Top IIα expression rates (P < 0.05). Whole-exome sequencing data analysis revealed that the mutational statuses of 373 genes (287 missense, 69 silent, 6 nonsense, and 11 synonymous mutations) differed significantly between PRCC and normal renal tissues (P < 0.05). Functional enrichment analysis was used to classify the 287 missense-mutated genes into 11 biological process clusters (comprised of 61 biological processes) and 5 pathways, involved in cell adhesion, microtubule-based movement, the cell cycle, polysaccharide biosynthesis, muscle cell development and differentiation, cell death, and negative regulation. Associated pathways included the ATP-binding cassette transporter, extracellular matrix-receptor interaction, lysosome, complement and coagulation cascades, and glyoxylate and dicarboxylate metabolism pathways. The missense mutation status of 19 genes differed significantly between the groups (P < 0.05), and alterations in the EEF1D, RFNG, GPR142, and RAB37 genes were located in different chromosomal regions in type 1 and 2 PRCC. These mutations may contribute to future studies on pathogenic mechanisms and targeted therapy of PRCC.
Collapse
Affiliation(s)
- Kunpeng Liu
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Yuan Ren
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Lijuan Pang
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Yan Qi
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Wei Jia
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Lin Tao
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Zhengyan Hu
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Jin Zhao
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Haijun Zhang
- Department of Pathology, First Affiliated Hospital of Medical School, Shihezi UniversityChina
| | - Li Li
- Department of Pathology, First Affiliated Hospital of Medical School, Shihezi UniversityChina
| | - Haifeng Yue
- Community hospital of Shihezi UniversityShihezi City, Xin Jiang, China
| | - Juan Han
- Community hospital of Shihezi UniversityShihezi City, Xin Jiang, China
| | - Weihua Liang
- Tongji Hospital Cancer Center, Tongji Medical College, Huazhong University of Science and TechnologyChina
| | - Jianming Hu
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Hong Zou
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| | - Xianglin Yuan
- Community hospital of Shihezi UniversityShihezi City, Xin Jiang, China
| | - Feng Li
- Department of Pathology, School of Medicine, Shihezi University, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaChina
| |
Collapse
|
34
|
Yan X, Xie J, Li J, Shuanghu C, Wu Z, Jian J. Screening and analysis on the protein interaction of the protein VP7 in grass carp reovirus. Virus Genes 2015; 50:425-33. [PMID: 25860999 DOI: 10.1007/s11262-015-1193-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 11/06/2014] [Indexed: 12/26/2022]
Abstract
Grass carp reovirus (GCRV) has caused serious economic losses for several decades in China. The protein VP7 is one of the important structural proteins in GCRV. Recent studies indicated that the protein VP7 had the commendable antigenicity and immunogenicity. The protein VP7 cooperated with VP5 could change the conformation of the cell membrane and facilitate entry of GCRV into host cells. We speculated that the protein VP7 should play an important role in the pathogenesis of GCRV. In order to explore the function of the protein VP7, the bait protein expression plasmid pGBKT7-vp7 and the cDNA library of CIK cells were constructed. By yeast two-hybrid system, after multiple screening with the high screening rate medium, rotary verification, sequencing and bioinformatics analysis, the interactions of the protein VP7 with ribosomal protein S20 (RPS20) and eukaryotic translation initiation factor 3 subunit b (eIF3b) in CIK cells were identified. RPS20 played the important roles in the generation of influenza B virus and a variety of diseases. eIF3b was relative to the infection of some viruses. This study suggested that the protein VP7 played the role in viral replication and most likely interacted with host proteins by RPS20 and eIF3b. The interaction mechanisms of the protein VP7 with RPS20 and eIF3b, and the subsequent effector mechanisms needed to be further studied. The corresponding protein interaction of the protein VP7 was not acquired in bioinformatics. The protein VP7 and its untranslated region may have the unknown special function. This study laid the foundation for deeply exploring the function of the protein VP7 in GCRV and had the important scientific significance for exploring the pathogenic mechanism of GCRV.
Collapse
Affiliation(s)
- Xiuying Yan
- Guangdong Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Guangdong Ocean University, Huguangyan East, Zhanjiang, 524088, China,
| | | | | | | | | | | |
Collapse
|
35
|
Veremieva M, Kapustian L, Khoruzhenko A, Zakharychev V, Negrutskii B, El'skaya A. Independent overexpression of the subunits of translation elongation factor complex eEF1H in human lung cancer. BMC Cancer 2014; 14:913. [PMID: 25472873 PMCID: PMC4265501 DOI: 10.1186/1471-2407-14-913] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/25/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The constituents of stable multiprotein complexes are known to dissociate from the complex to play independent regulatory roles. The components of translation elongation complex eEF1H (eEF1A, eEF1Bα, eEF1Bβ, eEF1Bγ) were found overexpressed in different cancers. To gain the knowledge about novel cancer-related translational mechanisms we intended to reveal whether eEF1H exists as a single unit or independent subunits in different human cancers. METHODS The changes in the expression level of every subunit of eEF1H in the human non-small-cell lung cancer tissues were examined. The localization of eEF1H subunits was assessed by immunohistochemistry methods, subcellular fractionation and confocal microscopy. The possibility of the interaction between the subunits was estimated by co-immunoprecipitation. RESULTS The level of eEF1Bβ expression was increased more than two-fold in 36%, eEF1Bγ in 28%, eEF1A in 20% and eEF1Bα in 8% of tumor specimens. The cancer-induced alterations in the subunits level were found to be uncoordinated, therefore the increase in the level of at least one subunit of eEF1H was observed in 52% of samples. Nuclear localization of eEF1Bβ in the cancer rather than distal normal looking tissues was found. In cancer tissue, eEF1A and eEF1Bα were not found in nuclei while all four subunits of eEF1H demonstrated both cytoplasmic and nuclear appearance in the lung carcinoma cell line A549. Unexpectedly, in the A549 nuclear fraction eEF1A lost the ability to interact with the eEF1B complex. CONCLUSIONS The results suggest independent functioning of some fraction of the eEF1H subunits in human tumors. The absence of eEF1A and eEF1B interplay in nuclei of A549 cells is a first evidence for non-translational role of nuclear-localized subunits of eEF1B. We conclude the appearance of the individual eEF1B subunits in tumors is a more general phenomenon than appreciated before and thus is a novel signal of cancer-related changes in translation apparatus.
Collapse
Affiliation(s)
| | | | | | | | - Boris Negrutskii
- State Key Laboratory of Molecular and Cellular Biology, Institute of Molecular Biology and Genetics NASU, 150 Acad,Zabolotnogo Str,, Kiev 03680, Ukraine.
| | | |
Collapse
|
36
|
Xie Y, Yang S, Cui X, Jiang L, Zhang S, Zhang Q, Zhang Y, Sun D. Identification and expression pattern of two novel alternative splicing variants of EEF1D gene of dairy cattle. Gene 2014; 534:189-96. [DOI: 10.1016/j.gene.2013.10.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/29/2013] [Accepted: 10/29/2013] [Indexed: 12/23/2022]
|
37
|
Tripaldi R, Stuppia L, Alberti S. Human height genes and cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:27-41. [PMID: 23428607 DOI: 10.1016/j.bbcan.2013.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/05/2013] [Accepted: 02/08/2013] [Indexed: 12/30/2022]
Abstract
Body development requires the ability to control cell proliferation and metabolism, together with selective 'invasive' cell migration for organogenesis. These requirements are shared with cancer. Human height-associated loci have been recently identified by genome-wide SNP-association studies. Strikingly, most of the more than 100 genes found associated to height appear linked to neoplastic growth, and impose a higher risk for cancer. Height-associated genes drive the HH/PTCH and BMP/TGFβ pathways, with p53, c-Myc, ERα, HNF4A and SMADs as central network nodes. Genetic analysis of body-size-affecting diseases and evidence from genetically-modified animals support this model. The finding that cancer is deeply linked to normal, body-plan master genes may profoundly affect current paradigms on tumor development.
Collapse
Affiliation(s)
- Romina Tripaldi
- Unit of Cancer Pathology, Department of Neuroscience and Imaging and CeSI, Foundation University G. d'Annunzio, Chieti, Italy
| | | | | |
Collapse
|
38
|
Zalatimo O, Zoccoli CM, Patel A, Weston CL, Glantz M. Impact of genetic targets on primary brain tumor therapy: what's ready for prime time? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:267-89. [PMID: 23288644 DOI: 10.1007/978-1-4614-6176-0_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Primary brain tumors constitute a substantial public health problem with 66,290 cases diagnosed in the US in 2012, and 13,700 deaths recorded. With discovery of genetic factors associated with specific brain tumor subtypes, the goal of therapy is changing from treating a class of tumors to developing individualized therapies catering to the molecular composition of the actual tumor. For oligodendrogliomas, the loss of 1p/19q due to an unbalanced translocation improves both survival and the response to therapy, and is thus both a prognostic and a predictive marker. Several additional genetic alterations such as EGFR amplification, MGMT methylation, PDGFR activation, and 9p and 10q loss, have improved our understanding of the characteristics of these tumors and may help guide therapy in the future. For astrocytic tumors, MGMT is associated with a better prognosis and an improved response to temozolomide, and for all glial tumors, mutations in the IDH1 gene are possibly the most potent of good prognostic markers. Three of these markers - 1p/19q deletions, MGMT methylation status, and mutations in the IDH1 gene - are so potent that a new brain tumor subtype, the "triple negative" glioma (1p/19q intact, MGMT unmethylated, IDH1 non-mutated) has entered common parlance. Newer markers, such as CD 133, require additional investigation to determine their prognostic and predictive utility. In medulloblastomas, markers of WNT activation, MYCC/MCYN amplification, and TrkC expression levels are reliable prognostic indicators, but do not yet drive specific treatment selection. Many other proposed markers, such as 17q gain, TP53 mutations, and hMOF protein expression show promise, but are not yet ready for prime time. In this chapter, we focus on the markers that have shown convincing prognostic, predictive, and diagnostic value, and discuss potential markers that are being currently being intensively investigated. We also discuss serum profiling of tumors in an effort to discover additional potential markers.
Collapse
Affiliation(s)
- O Zalatimo
- Department of Neurosurgery, Penn State College of Medicine, Hershey Medical Center, EC 1001, 30 Hope Drive, Hershey, PA 17033, USA.
| | | | | | | | | |
Collapse
|
39
|
Lau KM, Chan QKY, Pang JCS, Ma FMT, Li KKW, Yeung WW, Cheng ASL, Feng H, Chung NYF, Li HM, Zhou L, Wang Y, Mao Y, Ng HK. Overexpression of HMGA1 deregulates tumor growth via cdc25A and alters migration/invasion through a cdc25A-independent pathway in medulloblastoma. Acta Neuropathol 2012; 123:553-71. [PMID: 22249617 DOI: 10.1007/s00401-011-0934-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 12/23/2011] [Indexed: 11/24/2022]
Abstract
Overexpression of high mobility group AT-hook 1 (HMGA1) is common in human cancers. Little is known about the mechanisms underlying its deregulation and downstream targets, and information about its clinical and biological significance in medulloblastoma (MB) is lacking. Here, we demonstrated frequent genomic gain at 6p21.33-6p21.31 with copy number increase leading to overexpression of HMGA1 in MB. The overexpression correlated with a high proliferation index and poor prognosis. Moreover, we found that hsa-miR-124a targeted 3'UTR of HMGA1 and negatively modulated the expression in MB cells, indicating that loss/downregulation of hsa-miR-124a reported in our previous study could contribute to the overexpression. Regarding the biological significance of HMGA1, siRNA knockdown and ectopic expression studies revealed the crucial roles of HMGA1 in controlling MB cell growth and migration/invasion through modulation of apoptosis and formation of filopodia and stress fibers, respectively. Furthermore, we identified cdc25A as a target of HMGA1 and showed that physical interaction between HMGA1 and the cdc25A promoter is required for transcriptional upregulation. In clinical samples, HMGA1 and cdc25A were concordantly overexpressed. Functionally, cdc25A is involved in the HMGA1-mediated control of MB cell growth. Finally, netropsin, which competes with HMGA1 in DNA binding, reduced the expression of cdc25A by suppression of its promoter activity and inhibited in vitro and in vivo intracranial MB cell growth. In conclusion, our results delineate the mechanisms underlying the deregulation and reveal the functional significance of HMGA1 in controlling MB cell growth and migration/invasion. Importantly, the results highlight the therapeutic potential of targeting HMGA1 in MB patients.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Animals
- Antiviral Agents/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Movement/genetics
- Cell Proliferation/drug effects
- Cerebellar Neoplasms/genetics
- Cerebellar Neoplasms/metabolism
- Cerebellar Neoplasms/mortality
- Cerebellar Neoplasms/pathology
- Chromatin Immunoprecipitation
- Chromosome Aberrations
- Chromosomes, Human, Pair 17
- Chromosomes, Human, Pair 6
- Dose-Response Relationship, Drug
- Electrophoretic Mobility Shift Assay
- Female
- Flow Cytometry
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Gene Knockout Techniques
- HMGA1a Protein/genetics
- HMGA1a Protein/metabolism
- Humans
- Male
- Medulloblastoma/genetics
- Medulloblastoma/metabolism
- Medulloblastoma/mortality
- Medulloblastoma/pathology
- Mice
- Mice, Nude
- Neoplasm Invasiveness/pathology
- Netropsin/pharmacology
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Survival Analysis
- Time Factors
- Xenograft Model Antitumor Assays
- cdc25 Phosphatases/genetics
- cdc25 Phosphatases/metabolism
Collapse
Affiliation(s)
- Kin-Mang Lau
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tsai-Morris CH, Sato H, Gutti R, Dufau ML. Role of gonadotropin regulated testicular RNA helicase (GRTH/Ddx25) on polysomal associated mRNAs in mouse testis. PLoS One 2012; 7:e32470. [PMID: 22479328 PMCID: PMC3316541 DOI: 10.1371/journal.pone.0032470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/31/2012] [Indexed: 01/06/2023] Open
Abstract
Gonadotropin Regulated Testicular RNA Helicase (GRTH/Ddx25) is a testis-specific multifunctional RNA helicase and an essential post-transcriptional regulator of spermatogenesis. GRTH transports relevant mRNAs from nucleus to cytoplasmic sites of meiotic and haploid germ cells and associates with actively translating polyribosomes. It is also a negative regulator of steroidogenesis in Leydig cells. To obtain a genome-wide perspective of GRTH regulated genes, in particularly those associated with polyribosomes, microarray differential gene expression analysis was performed using polysome-bound RNA isolated from testes of wild type (WT) and GRTH KO mice. 792 genes among the entire mouse genome were found to be polysomal GRTH-linked in WT. Among these 186 were down-regulated and 7 up-regulated genes in GRTH null mice. A similar analysis was performed using total RNA extracted from purified germ cell populations to address GRTH action in individual target cells. The down-regulation of known genes concerned with spermatogenesis at polysomal sites in GRTH KO and their association with GRTH in WT coupled with early findings of minor or unchanged total mRNAs and abolition of their protein expression in KO underscore the relevance of GRTH in translation. Ingenuity pathway analysis predicted association of GRTH bound polysome genes with the ubiquitin-proteasome-heat shock protein signaling network pathway and NFκB/TP53/TGFB1 signaling networks were derived from the differentially expressed gene analysis. This study has revealed known and unexplored factors in the genome and regulatory pathways underlying GRTH action in male reproduction.
Collapse
Affiliation(s)
- Chon-Hwa Tsai-Morris
- Section on Molecular Endocrinology, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | |
Collapse
|
41
|
Gyenis L, Duncan JS, Turowec JP, Bretner M, Litchfield DW. Unbiased functional proteomics strategy for protein kinase inhibitor validation and identification of bona fide protein kinase substrates: application to identification of EEF1D as a substrate for CK2. J Proteome Res 2011; 10:4887-901. [PMID: 21936567 PMCID: PMC3208357 DOI: 10.1021/pr2008994] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Protein kinases have emerged as attractive targets for treatment of several diseases prompting large-scale phosphoproteomics studies to elucidate their cellular actions and the design of novel inhibitory compounds. Current limitations include extensive reliance on consensus predictions to derive kinase-substrate relationships from phosphoproteomics data and incomplete experimental validation of inhibitors. To overcome these limitations in the case of protein kinase CK2, we employed functional proteomics and chemical genetics to enable identification of physiological CK2 substrates and validation of CK2 inhibitors including TBB and derivatives. By 2D electrophoresis and mass spectrometry, we identified the translational elongation factor EEF1D as a protein exhibiting CK2 inhibitor-dependent decreases in phosphorylation in (32)P-labeled HeLa cells. Direct phosphorylation of EEF1D by CK2 was shown by performing CK2 assays with EEF1D -FLAG from HeLa cells. Dramatic increases in EEF1D phosphorylation following λ-phosphatase treatment and phospho- EEF1D antibody recognizing EEF1D pS162 indicated phosphorylation at the CK2 site in cells. Furthermore, phosphorylation of EEF1D in the presence of TBB or TBBz is restored using CK2 inhibitor-resistant mutants. Collectively, our results demonstrate that EEF1D is a bona fide physiological CK2 substrate for CK2 phosphorylation. Furthermore, this validation strategy could be adaptable to other protein kinases and readily combined with other phosphoproteomic methods.
Collapse
Affiliation(s)
- Laszlo Gyenis
- Department of Biochemistry, The University of Western Ontario , London, Ontario, N6A 5C1, Canada
| | | | | | | | | |
Collapse
|
42
|
Zuccotti M, Merico V, Bellone M, Mulas F, Sacchi L, Rebuzzini P, Prigione A, Redi CA, Bellazzi R, Adjaye J, Garagna S. Gatekeeper of pluripotency: a common Oct4 transcriptional network operates in mouse eggs and embryonic stem cells. BMC Genomics 2011; 12:1-13. [PMID: 21729306 PMCID: PMC3154874 DOI: 10.1186/1471-2164-12-345] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 07/05/2011] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Oct4 is a key factor of an expanded transcriptional network (Oct4-TN) that governs pluripotency and self-renewal in embryonic stem cells (ESCs) and in the inner cell mass from which ESCs are derived. A pending question is whether the establishment of the Oct4-TN initiates during oogenesis or after fertilisation. To this regard, recent evidence has shown that Oct4 controls a poorly known Oct4-TN central to the acquisition of the mouse egg developmental competence. The aim of this study was to investigate the identity and extension of this maternal Oct4-TN, as much as whether its presence is circumscribed to the egg or maintained beyond fertilisation. RESULTS By comparing the genome-wide transcriptional profile of developmentally competent eggs that express the OCT4 protein to that of developmentally incompetent eggs in which OCT4 is down-regulated, we unveiled a maternal Oct4-TN of 182 genes. Eighty of these transcripts escape post-fertilisation degradation and represent the maternal Oct4-TN inheritance that is passed on to the 2-cell embryo. Most of these 80 genes are expressed in cancer cells and 37 are notable companions of the Oct4 transcriptome in ESCs. CONCLUSIONS These results provide, for the first time, a developmental link between eggs, early preimplantation embryos and ESCs, indicating that the molecular signature that characterises the ESCs identity is rooted in oogenesis. Also, they contribute a useful resource to further study the mechanisms of Oct4 function and regulation during the maternal-to-embryo transition and to explore the link between the regulation of pluripotency and the acquisition of de-differentiation in cancer cells.
Collapse
Affiliation(s)
- Maurizio Zuccotti
- Sezione di Istologia ed Embriologia, Dipartimento di Medicina Sperimentale, Universita' degli Studi di Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhang MX, Zhang YM, Esther J, He W, Che HH, Gu P. Effects of yiqi huoxue recipe and Coxsackie virus B type 3 on the expression of ribosomal protein S20 in rat cardiac myocytes. Chin J Integr Med 2011; 17:376-80. [PMID: 21611902 DOI: 10.1007/s11655-011-0729-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To study the effects of yiqi huoxue recipe and Coxsackie B virus type 3 (CVB3) on the expression of ribosomal protein S20 in rat cardiac myocytes, to explore the pathogenesis of myocarditis induced by CVB3 and the mechanism of yiqi huoxue recipe on gene level, and to further investigate whether yiqi huoxue recipe is an effective prescription for CVB3 myocarditis. METHODS A modified suppression subtractive hybridization (SSH) was used to isolate differentially expressed genes between the CVB3 infection group and the treatment group with yiqi huoxue recipe. The results were further verified by fluorescence RT-PCR. RESULTS The results of SSH showed that the gene expression of ribosomal protein S20 in the treatment group was higher than that in the CVB3 infection group (P<0.05), which agreed with the results of fluorescent RT-PCR. CONCLUSION Down-regulation of ribosomal protein S20 mRNA expression might be one of the mechanisms in CVB3 myocarditis, and yiqi huoxue recipe could achieve the treatment of viral myocarditis by regulating the expression of ribosomal protein S20.
Collapse
Affiliation(s)
- Ming-xue Zhang
- Department of Traditional Chinese Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China.
| | | | | | | | | | | |
Collapse
|
44
|
Veremieva M, Khoruzhenko A, Zaicev S, Negrutskii B, El'skaya A. Unbalanced expression of the translation complex eEF1 subunits in human cardioesophageal carcinoma. Eur J Clin Invest 2011; 41:269-76. [PMID: 20964681 DOI: 10.1111/j.1365-2362.2010.02404.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The signalling role of individual subunits released from some stable translation multi-molecular complexes under unfavourable circumstances is known. The disease-related role of the translation elongation factor 1 complex (eEF1) as a whole is never researched; however, its subunits possess apparent regulatory potency. Whether the individual eEF1 subunits can exist and function in cell beyond the complex is not known. MATERIALS AND METHODS The protein and mRNA levels of the A1, Bα, Bβ or Bγ subunits of eEF1 were analysed by Western and Northern blot techniques in the same specimens of cardioesophageal carcinoma and correspondingly paired normal tissues. Cancer-induced changes in localization patterns of the eEF1 subunits were examined immunohistochemically. RESULTS Changes in different eEF1 subunits expression were found to be unbalanced, indicating cancer-related emergence of individual components of the eEF1 complex. Independent overexpression of at least one eEF1 component was observed in 72% clinical samples. Noncomplexed eEF1B subunits were also detected by immunohistochemical analysis. In the normal tissue, localization of the Bα, Bβ and Bγ subunits was nuclear-cytoplasmic while in the cancer tissue the only Bγ subunit stayed in nucleus. CONCLUSIONS Our data are first to indicate that the individual subunits can exist separately from the eEF1B complex in cancer tissues and that disintegration of eEF1B could be an important sign of cancer development. Nuclear localization of Bγ both in normal and in cancer tissues suggests its previously unknown nucleus-specific role in human cells.
Collapse
Affiliation(s)
- Marina Veremieva
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | | | | | | | | |
Collapse
|
45
|
Abstract
Despite the fact that ribosomal proteins are the constituents of an organelle that is present in every cell, they show a surprising level of regulation, and several of them have also been shown to have other extra-ribosomal functions, such in replication, transcription, splicing or even ageing. This review provides a comprehensive summary of these important aspects.
Collapse
Affiliation(s)
- Rital B Bhavsar
- Department of Biology, University of Dayton, OH 45469-2320, USA
| | | | | |
Collapse
|
46
|
McCabe MG, Bäcklund LM, Leong HS, Ichimura K, Collins VP. Chromosome 17 alterations identify good-risk and poor-risk tumors independently of clinical factors in medulloblastoma. Neuro Oncol 2011; 13:376-83. [PMID: 21292688 PMCID: PMC3064691 DOI: 10.1093/neuonc/noq192] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Current risk stratification schemas for medulloblastoma, based on combinations of clinical variables and histotype, fail to accurately identify particularly good- and poor-risk tumors. Attempts have been made to improve discriminatory power by combining clinical variables with cytogenetic data. We report here a pooled analysis of all previous reports of chromosomal copy number related to survival data in medulloblastoma. We collated data from previous reports that explicitly quoted survival data and chromosomal copy number in medulloblastoma. We analyzed the relative prognostic significance of currently used clinical risk stratifiers and the chromosomal aberrations previously reported to correlate with survival. In the pooled dataset metastatic disease, incomplete tumor resection and severe anaplasia were associated with poor outcome, while young age at presentation was not prognostically significant. Of the chromosomal variables studied, isolated 17p loss and gain of 1q correlated with poor survival. Gain of 17q without associated loss of 17p showed a trend to improved outcome. The most commonly reported alteration, isodicentric chromosome 17, was not prognostically significant. Sequential multivariate models identified isolated 17p loss, isolated 17q gain, and 1q gain as independent prognostic factors. In a historical dataset, we have identified isolated 17p loss as a marker of poor outcome and 17q gain as a novel putative marker of good prognosis. Biological markers of poor-risk and good-risk tumors will be critical in stratifying treatment in future trials. Our findings should be prospectively validated independently in future clinical studies.
Collapse
Affiliation(s)
- Martin G McCabe
- Manchester Academic Health Science Centre, School of Cancer and Enabling Sciences, University of Manchester, The Christie NHS Foundation Trust, Withington, Manchester M20 4BX, UK.
| | | | | | | | | |
Collapse
|
47
|
Zakrzewska M, Zakrzewski K, Grešner SM, Piaskowski S, Zalewska-Szewczyk B, Liberski PP. Polycomb genes expression as a predictor of poor clinical outcome in children with medulloblastoma. Childs Nerv Syst 2011; 27:79-86. [PMID: 20717685 PMCID: PMC3015167 DOI: 10.1007/s00381-010-1260-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 07/30/2010] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Medulloblastoma is the most frequent type of embryonal tumor in the pediatric population, accounting for 20-25% of all brain tumors in children. Recently, the suspected contribution of the Polycomb group (PcG) genes in medulloblastoma development was described. PcG genes play an important role in developmental processes; they are also involved in the self-renewal of hematopoietic and neural stem cells as well as in malignant transformation. PURPOSE In this study, we evaluated the expression of BMI1and PCGF2, members of family of PcG genes, and their potential target, MYC oncogene, and analyzed their association with demographic and clinical data. MATERIALS AND METHODS Thirty-one children (18 males and 13 females, aged from 0.4 to 17 years) with medulloblastoma were included in this study. The gene's expression level was measured by quantitative real-time PCR, obtained using the two-color multiplexing technique. RESULTS We found that the higher expression levels of BMI1 and PCGF2 genes were associated with significantly decreased patient survival (p = 0.02 and p = 0.012, respectively). Significant differences between gender were found, with a higher expression level of the PCGF2 gene observed among females (p = 0.02). CONCLUSION Our analysis showed correlation between BMI1 and PCGF2 gene's expression and survival in children with medulloblastoma.
Collapse
Affiliation(s)
- Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Łódź, Czechosłowacka 8/10, Łódź, Poland.
| | - Krzysztof Zakrzewski
- Department of Neurosurgery, Polish Mother’s Memorial Hospital Research Institute, Rzgowska 281/289, 93-338 Łódź, Poland
| | - Sylwia M. Grešner
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Łódź, Czechosłowacka 8/10, 92-216 Łódź, Poland
| | - Sylwester Piaskowski
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Łódź, Czechosłowacka 8/10, 92-216 Łódź, Poland
| | - Beata Zalewska-Szewczyk
- Department of Pediatrics, Oncology, Hematology and Diabetology, 1st Chair of Pediatrics, Medical University of Łódź, Sporna 36/50, 91-738 Łódź, Poland
| | - Paweł P. Liberski
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Łódź, Czechosłowacka 8/10, 92-216 Łódź, Poland
| |
Collapse
|
48
|
Minichromosome maintenance proteins 2, 3 and 7 in medulloblastoma: overexpression and involvement in regulation of cell migration and invasion. Oncogene 2010; 29:5475-89. [DOI: 10.1038/onc.2010.287] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
49
|
Veremieva MV, Malysheva TA, Zozulya YP, Rozumenko VD, Sidorik LL, Kavsan VM, Negrutskii BS, El'skaya AV. Multisubunit complex eEF1H in human glial tumors: from mRNA to protein. ACTA ACUST UNITED AC 2010. [DOI: 10.7124/bc.000166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- M. V. Veremieva
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - T. A. Malysheva
- Institute of neurosurgery named after A. P. Romodanov, AMS of Ukraine
| | - Y. P. Zozulya
- Institute of neurosurgery named after A. P. Romodanov, AMS of Ukraine
| | - V. D. Rozumenko
- Institute of neurosurgery named after A. P. Romodanov, AMS of Ukraine
| | - L. L. Sidorik
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - V. M. Kavsan
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - B. S. Negrutskii
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| | - A. V. El'skaya
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine
| |
Collapse
|
50
|
Yu L, Guan YJ, Gao Y, Wang X. Rpl30 and Hmgb1 are required for neurulation in golden hamster. Int J Neurosci 2010; 119:1076-90. [PMID: 19922340 DOI: 10.1080/00207450802330504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neural tube defects (NTDs) are a group of severe congenital malformations resulting from the failure of neurulation. Genes influencing neurulation have been investigated for their contribution to NTDs. Ribosomal protein (Rp) is an abundant and belongs to a high conservative gene family, which has the complex task of coordinating protein biosynthesis in order to maintain cell homeostasis and survival. However, the mechanisms of Rp in the NTDs are unknown. Understanding the mechanisms will lead to new insights into NTDs. In this report, we constructed a cDNA library from neural tube of golden hamster and screened the cDNA library by a subsection screening method (SSS). Our results demonstrate a possible essential role of the RPL30 cDNA gene during neurulation and in the risk of NTDs. Our study also suggests that another gene, HMGB1, may be significantly associated with neurulation and the risk of NTDs.
Collapse
Affiliation(s)
- Li Yu
- Department of Histology and Embryology, Weifang Medical College, Weifang, China
| | | | | | | |
Collapse
|