1
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
2
|
Hua J, Wang K, Chen Y, Xu X, Dong G, Li Y, Liu R, Xiong Y, Ding J, Zhang T, Zeng X, Li Y, Sun H, Gu Y, Liu S, Ouyang W, Liu C. Molecular characterization of human HSPCs with different cell fates in vivo using single-cell transcriptome analysis and lentiviral barcoding technology. Clin Transl Med 2024; 14:e70085. [PMID: 39538416 PMCID: PMC11560861 DOI: 10.1002/ctm2.70085] [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: 07/04/2024] [Revised: 10/17/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) possess the potential to produce all types of blood cells throughout their lives. It is well recognized that HSPCs are heterogeneous, which is of great significance for their clinical applications and the treatment of diseases associated with HSPCs. This study presents a novel technology called Single-Cell transcriptome Analysis and Lentiviral Barcoding (SCALeBa) to investigate the molecular mechanisms underlying the heterogeneity of human HSPCs in vivo. The SCALeBa incorporates a transcribed barcoding library and algorithm to analyze the individual cell fates and their gene expression profiles simultaneously. Our findings using SCALeBa reveal that HSPCs subset with stronger stemness highly expressed MYL6B, ATP2A2, MYO19, MDN1, ING3, and so on. The high expression of COA3, RIF1, RAB14, and GOLGA4 may contribute to the pluripotent-lineage differentiation of HSPCs. Moreover, the roles of the representative genes revealed in this study regarding the stemness of HPSCs were confirmed with biological experiments. HSPCs expressing MRPL23 and RBM4 genes may contribute to differentiation bias into myeloid and lymphoid lineage, respectively. In addition, transcription factor (TF) characteristics of lymphoid and myeloid differentiation bias HSPCs subsets were identified and linked to previously identified genes. Furthermore, the stemness, pluripotency, and differentiation-bias genes identified with SCALeBa were verified in another independent HSPCs dataset. Finally, this study proposes using the SCALeBa-generated tracking trajectory to improve the accuracy of pseudo-time analysis results. In summary, our study provides valuable insights for understanding the heterogeneity of human HSPCs in vivo and introduces a novel technology, SCALeBa, which holds promise for broader applications. KEY POINTS: SCALeBa and its algorithm are developed to study the molecular mechanism underlying human HSPCs identity and function. The human HSPCs expressing MYL6B, MYO19, ATP2A2, MDN1, ING3, and PHF20 may have the capability for high stemness. The human HSPCs expressing COA3, RIF1, RAB14, and GOLGA4 may have the capability for pluripotent-lineage differentiation. The human HSPCs expressing MRPL23 and RBM4 genes may have the capability to differentiate into myeloid and lymphoid lineage respectively in vivo. The legitimacy of the identified genes with SCALeBa was validated using biological experiments and a public human HSPCs dataset. SCALeBa improves the accuracy of differentiation trajectories in monocle2-based pseudo-time analysis.
Collapse
Affiliation(s)
- Junnan Hua
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGIShenzhenChina
| | - Ke Wang
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | | | - Xiaojing Xu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGIShenzhenChina
| | - Guoyi Dong
- BGIShenzhenChina
- BGI Hemogen TherapeuticShenzhenChina
| | - Yue Li
- Department of Hematology and OncologyShenzhen Children's HospitalShenzhenChina
| | - Rui Liu
- BGIShenzhenChina
- Department of Hematology and OncologyShenzhen Children's HospitalShenzhenChina
| | - Yecheng Xiong
- BGIShenzhenChina
- BGI Hemogen TherapeuticShenzhenChina
| | - Jiabin Ding
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGIShenzhenChina
| | | | - Xinru Zeng
- BGIShenzhenChina
- BGI Hemogen TherapeuticShenzhenChina
| | | | | | | | - Sixi Liu
- Department of Hematology and OncologyShenzhen Children's HospitalShenzhenChina
| | - Wenjie Ouyang
- BGIShenzhenChina
- BGI Hemogen TherapeuticShenzhenChina
| | - Chao Liu
- BGIShenzhenChina
- BGI Hemogen TherapeuticShenzhenChina
| |
Collapse
|
3
|
Zhang F, Xiong Q, Wang M, Cao X, Zhou C. FUBP1 in human cancer: Characteristics, functions, and potential applications. Transl Oncol 2024; 48:102066. [PMID: 39067088 PMCID: PMC11338137 DOI: 10.1016/j.tranon.2024.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/04/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Far upstream element-binding protein 1 (FUBP1) is a single-stranded nucleic acid-binding protein that binds to the Far Upstream Element (FUSE) sequence and is involved in important biological processes, including DNA transcription, RNA biogenesis, and translation. Recent studies have highlighted the significance of aberrant expression or mutations in FUBP1 in the development of various tumors, with FUBP1 overexpression often indicating oncogenic roles in different tumor types. However, it is worth noting that recent research has discovered its tumor-suppressive role in cancer, which is not yet fully understood and appears to be tissue- or context-dependent. This review summarizes the association between FUBP1 and diverse cancers and discusses the functions of FUBP1 in cancer. In addition, this review proposes potential clinical implications and outlines future research directions to pave the way for the development of targeted therapeutic strategies focusing on FUBP1.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, No 256 Youyi West Road, Xi'an, 710068, Shaanxi, China
| | - Qunli Xiong
- Department of Abdominal Oncology, West China Hospital, Sichuan University, No 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Min Wang
- Department of Science and Education, Xi'an Children's Hospital Affiliated of Xi'an Jiaotong University, No 69 Xijuyuan lane, Xi'an, 710002, Shaanxi, China
| | - Ximing Cao
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, No 256 Youyi West Road, Xi'an, 710068, Shaanxi, China
| | - Congya Zhou
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, No 256 Youyi West Road, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
4
|
Dicanio M, Giaccherini M, Clay‐Gilmour A, Macauda A, Sainz J, Machiela MJ, Rybicka‐Ramos M, Norman AD, Tyczyńska A, Chanock SJ, Barington T, Kumar SK, Bhatti P, Cozen W, Brown EE, Suska A, Haastrup EK, Orlowski RZ, Dudziński M, Garcia‐Sanz R, Kruszewski M, Martinez‐Lopez J, Beider K, Iskierka‐Jazdzewska E, Pelosini M, Berndt SI, Raźny M, Jamroziak K, Rajkumar SV, Jurczyszyn A, Vangsted AJ, Collado PG, Vogel U, Hofmann JN, Petrini M, Butrym A, Slager SL, Ziv E, Subocz E, Giles GG, Andersen NF, Mazur G, Watek M, Lesueur F, Hildebrandt MAT, Zawirska D, Ebbesen LH, Marques H, Gemignani F, Dumontet C, Várkonyi J, Buda G, Nagler A, Druzd‐Sitek A, Wu X, Kadar K, Camp NJ, Grzasko N, Waller RG, Vachon C, Canzian F, Campa D. A pleiotropic variant in DNAJB4 is associated with multiple myeloma risk. Int J Cancer 2023; 152:239-248. [PMID: 36082445 PMCID: PMC9828677 DOI: 10.1002/ijc.34278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 01/12/2023]
Abstract
Pleiotropy, which consists of a single gene or allelic variant affecting multiple unrelated traits, is common across cancers, with evidence for genome-wide significant loci shared across cancer and noncancer traits. This feature is particularly relevant in multiple myeloma (MM) because several susceptibility loci that have been identified to date are pleiotropic. Therefore, the aim of this study was to identify novel pleiotropic variants involved in MM risk using 28 684 independent single nucleotide polymorphisms (SNPs) from GWAS Catalog that reached a significant association (P < 5 × 10-8 ) with their respective trait. The selected SNPs were analyzed in 2434 MM cases and 3446 controls from the International Lymphoma Epidemiology Consortium (InterLymph). The 10 SNPs showing the strongest associations with MM risk in InterLymph were selected for replication in an independent set of 1955 MM cases and 1549 controls from the International Multiple Myeloma rESEarch (IMMEnSE) consortium and 418 MM cases and 147 282 controls from the FinnGen project. The combined analysis of the three studies identified an association between DNAJB4-rs34517439-A and an increased risk of developing MM (OR = 1.22, 95%CI 1.13-1.32, P = 4.81 × 10-7 ). rs34517439-A is associated with a modified expression of the FUBP1 gene, which encodes a multifunctional DNA and RNA-binding protein that it was observed to influence the regulation of various genes involved in cell cycle regulation, among which various oncogenes and oncosuppressors. In conclusion, with a pleiotropic scan approach we identified DNAJB4-rs34517439 as a potentially novel MM risk locus.
Collapse
Affiliation(s)
| | | | - Alyssa Clay‐Gilmour
- Department of Epidemiology and Biostatistics, Arnold School of Public HealthUniversity of South CarolinaGreenvilleSouth CarolinaUSA
| | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Juan Sainz
- Genomic Oncology Area, GENYO. Center for Genomics and Oncological Research: PfizerUniversity of Granada/Andalusian Regional GovernmentGranadaSpain,Department of HematologyVirgen de las Nieves University HospitalGranadaSpain,Department of MedicineUniversity of GranadaGranadaSpain
| | - Mitchell J. Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institues of HealthBethesdaMarylandUSA
| | | | - Aaron D. Norman
- Division of Epidemiology, Department of Health Sciences ResearchMayo ClinicRochesterOntarioUSA,Division of Biomedical Statistics and Informatics, Department of Health Sciences ResearchMayo ClinicRochesterOntarioUSA
| | - Agata Tyczyńska
- Department of Hematology and TransplantologyMedical University of GdańskGdańskPoland
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institues of HealthBethesdaMarylandUSA
| | | | - Shaji K. Kumar
- Division of Hematology, Department of Internal MedicineMayo ClinicRochesterOntarioUSA
| | - Parveen Bhatti
- Cancer Control ResearchBC CancerVancouverCanada,Program in Epidemiology, Public Health Sciences DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Wendy Cozen
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, Susan and Henry Samueli College of Health SciencesChao Family Comprehensive Cancer Center, University of CaliforniaIrvineCaliforniaUSA,Department of Pathology, School of Medicine, Susan and Henry Samueli College of Health SciencesChao Family Comprehensive Cancer Center, University of CaliforniaIrvineCaliforniaUSA
| | - Elizabeth E. Brown
- Department of Pathology, School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anna Suska
- Plasma Cell Dyscrasia Center Department of Hematology Jagiellonian University Faculty of MedicineKrakówPoland
| | | | - Robert Z. Orlowski
- Department of Lymphoma ‐ Myeloma, Division of Cancer MedicineUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Marek Dudziński
- Department of Hematology, Institute of Medical Sciences, College of Medical SciencesUniversity of RzeszowRzeszowPoland
| | - Ramon Garcia‐Sanz
- Medina A. Department of Hematology, University Hospital of Salamanca (HUS/IBSAL)CIBERONC and Cancer Research Institute of Salamanca‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Marcin Kruszewski
- Department of HematologyUniversity Hospital No. 2 in BydgoszczBydgoszczPoland
| | | | - Katia Beider
- Hematology Division Chaim Sheba Medical CenterTel HashomerIsrael
| | | | - Matteo Pelosini
- U.O. Dipartimento di EmatologiaAzienda USL Toscana Nord OvestLivornoItaly,Present address:
Ospedale Santa ChiaraPisaItaly
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institues of HealthBethesdaMarylandUSA
| | | | - Krzysztof Jamroziak
- Department of HematologyInstitute of Hematology and Transfusion MedicineWarsawPoland
| | - S. Vincent Rajkumar
- Division of Hematology, Department of Internal MedicineMayo ClinicRochesterOntarioUSA
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasia Center Department of Hematology Jagiellonian University Faculty of MedicineKrakówPoland
| | | | | | - Ulla Vogel
- National Research Center for the Working EnvironmentCopenhagenDenmark
| | - Jonathan N. Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institues of HealthBethesdaMarylandUSA
| | - Mario Petrini
- Hematology Unit, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Aleksandra Butrym
- Department of Cancer Prevention and TherapyWroclaw Medical UniversityWroclawPoland
| | - Susan L. Slager
- Division of Epidemiology, Department of Health Sciences ResearchMayo ClinicRochesterOntarioUSA
| | - Elad Ziv
- Department of MedicineUniversity of California San Francisco Helen Diller Family Comprehensive Cancer CenterSan FranciscoCaliforniaUSA
| | - Edyta Subocz
- Department of HematologyMilitary Institute of MedicineWarsawPoland
| | - Graham G. Giles
- Cancer Epidemiology DivisionCancer Council VictoriaMelbourneVictoriaAustralia,Center for Epidemiology and Biostatistics, School of Population and Global HealthThe University of MelbourneMelbourneVictoriaAustralia,Precision Medicine, School of Clinical Sciences at Monash HealthMonash UniversityClaytonVictoriaAustralia
| | | | - Grzegorz Mazur
- Department of Internal Diseases, Occupational Medicine, Hypertension and Clinical OncologyWroclaw Medical UniversityWroclawPoland
| | - Marzena Watek
- Department of HematologyInstitute of Hematology and Transfusion MedicineWarsawPoland,Department of HematologyHolycross Cancer CenterKielcePoland
| | - Fabienne Lesueur
- Inserm, U900, Institut Curie, PSL Research University, Mines ParisTechParisFrance
| | - Michelle A. T. Hildebrandt
- Department of Lymphoma ‐ Myeloma, Division of Cancer MedicineUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Daria Zawirska
- Department of HematologyUniversity Hospital in CracowCracowPoland
| | | | - Herlander Marques
- Life and Health Sciences Research Institute (ICVS), School of Health SciencesUniversity of Minho, Braga, Portugal and ICVS/3B's – PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | | | | | - Judit Várkonyi
- Department of Hematology and Internal MedicineSemmelweis UniversityBudapestHungary
| | - Gabriele Buda
- Hematology Unit, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Arnon Nagler
- Hematology Division Chaim Sheba Medical CenterTel HashomerIsrael
| | - Agnieszka Druzd‐Sitek
- Department of Lymphoproliferative DiseasesMaria Skłodowska‐Curie National Research Institute of OncologyWarsawPoland
| | - Xifeng Wu
- Department of Epidemiology, Division of Cancer Prevention and Population SciencesUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Katalin Kadar
- Department of Hematology and Internal MedicineSemmelweis UniversityBudapestHungary
| | - Nicola J. Camp
- Division of Hematology and Huntsman Cancer InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - Norbert Grzasko
- Department of Experimental HematooncologyMedical University of LublinLublinPoland
| | - Rosalie G. Waller
- Division of Biomedical Statistics and Informatics, Department of Health Sciences ResearchMayo ClinicRochesterOntarioUSA
| | - Celine Vachon
- Division of Epidemiology, Department of Health Sciences ResearchMayo ClinicRochesterOntarioUSA
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ)HeidelbergGermany
| | | |
Collapse
|
5
|
Yu W, Schmachtel T, Fawaz M, Rieger MA. Isolation of murine bone marrow hematopoietic stem and progenitor cell populations via flow cytometry. Methods Cell Biol 2022; 171:173-195. [DOI: 10.1016/bs.mcb.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
6
|
Yin H, Gao T, Xie J, Huang Z, Zhang X, Yang F, Qi W, Yang Z, Zhou T, Gao G, Yang X. FUBP1 promotes colorectal cancer stemness and metastasis via DVL1-mediated activation of Wnt/β-catenin signaling. Mol Oncol 2021; 15:3490-3512. [PMID: 34288405 PMCID: PMC8637553 DOI: 10.1002/1878-0261.13064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/02/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
Distant metastasis is, unfortunately, the leading cause of death in colorectal cancer (CRC). Approximately 50% of CRC patients develop liver metastases, while 10–30% of patients develop pulmonary metastases. The occurrence of metastasis is considered to be almost exclusively driven by cancer stem cells (CSCs) formation. However, the key molecules that confer the transformation to stem cells in CRC, and subsequent metastasis, remain unclear. Far upstream element‐binding protein 1 (FUBP1), a transcriptional regulator of c‐Myc, was screened in CSCs of CRC by mass spectrometry and was examined by immunohistochemistry in a cohort of CRC tissues. FUBP1 was upregulated in 85% of KRAS‐mutant and 25% of wild‐type CRC patients. Further, whether in KRAS‐mutant or wild‐type patients, elevated FUBP1 was positively correlated with CRC lymph node metastasis and clinical stage, and negatively associated with overall survival. Overexpression of FUBP1 significantly enhanced CRC cell migration, invasion, tumor sphere formation, and CD133 and ALDH1 expression in vitro, and tumorigenicity in vivo. Mechanistically, FUBP1 promoted the initiation of CSCs by activating Wnt/β‐catenin signaling via directly binding to the promoter of DVL1, a potent activator of β‐catenin. Knockdown of DVL1 significantly inhibited the transformation to stem cells in, as well as the tumorigenicity of, CRC. Activation of Wnt/β‐catenin signaling by DVL1 increased pluripotent transcription factors, including c‐Myc, NANOG, and SOX2. Moreover, FUBP1 was upregulated at the post‐transcriptional level. Elevated FUBP1 levels in KRAS wild‐type CRC patients is due to the decrease in Smurf2, which promotes ubiquitin‐mediated degradation of FUBP1. In contrast, FUBP1 was upregulated in KRAS‐mutant patients through both inhibition of caspase 3‐dependent cleavage and decreased Smurf2. Our results demonstrate, for the first time, that FUBP1 is an oncogene, initiating the development of CSCs, as well as a new powerful endogenous Wnt‐signaling agonist that could provide an important prognostic factor and therapeutic target for metastasis in both KRAS‐mutant and wild‐type CRC.
Collapse
Affiliation(s)
- Haofan Yin
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tianxiao Gao
- Department of Biochemistry, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, China
| | - Jinye Xie
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhijian Huang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoyan Zhang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fengyu Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weiwei Qi
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhonghan Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ti Zhou
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guoquan Gao
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| | - Xia Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
7
|
Zhang Y, Chen J, Zhou N, Lu Y, Lu J, Xing X, Chen H, Zhang X. FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma. Int J Mol Med 2021; 47:66. [PMID: 33649780 PMCID: PMC7952245 DOI: 10.3892/ijmm.2021.4899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/29/2021] [Indexed: 11/06/2022] Open
Abstract
Recent studies have reported that the expression levels of far upstream element‑binding protein 1 (FUBP1) were upregulated and served a crucial role in several types of cancer. However, the underlying molecular mechanisms and clinical significance of FUBP1 in pancreatic adenocarcinoma (PAAD) remain unclear. The present study aimed to determine the expression levels of FUBP1 in patients with PAAD and subsequently investigated the biological functions and mechanisms of FUBP1 using in vitro assays. FUBP1 expression levels and survival outcomes in patients with PAAD were analyzed using The Cancer Genome Atlas and starBase databases. Reverse transcription‑quantitative PCR was used to analyze the mRNA expression levels of FUBP1 in PAAD and adjacent normal tissues. In addition, the expression of FUBP1 was knocked down with small interfering RNA and overexpressed using FUBP1‑overexpressed plasmids, and the effects on biological functions, including cell proliferation, migration and invasion, were investigated. Western blotting and immunofluorescence assays were used to determine the role of FUBP1 in epithelial‑mesenchymal transition (EMT). The results of the present study revealed that the expression levels of FUBP1 were upregulated in PAAD tissues compared with adjacent normal tissues and the upregulated expression was significantly associated with poor survival. The knockdown of FUBP1 expression significantly inhibited the proliferative, migratory and invasive abilities of the PAAD PaTu8988 cell line, while the overexpression of FUBP1 promoted cell proliferation, migration and invasion in the PAAD SW1990 cell line. Furthermore, the knockdown of FUBP1 downregulated the expression levels of EMT‑related markers, including N‑cadherin, β‑catenin and vimentin, while the expression levels of E‑cadherin were upregulated. The knockdown of FUBP1 was also revealed to regulate the TGFβ/Smad signaling cascade by downregulating phosphorylated‑Smad2/3 and TGFβ1 expression levels. Conversely, the overexpression of FUBP1 reversed these effects. In conclusion, the findings of the present study indicated that FUBP1 may be a potential oncogene that mediates the EMT of PAAD via TGFβ/Smad signaling. These data suggested that FUBP1 may represent a potential biomarker for the diagnosis of PAAD or a target for the treatment of patients with PAAD.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai 201499, P.R. China
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Jinlian Chen
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai 201499, P.R. China
| | - Nvshi Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yun Lu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Jingwen Lu
- Department of Pathology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Xin Xing
- Central Laboratory, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai 201499, P.R. China
| | - Hua Chen
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai 201499, P.R. China
| | - Xingxing Zhang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai 201499, P.R. China
| |
Collapse
|
8
|
Zheng Y, Dubois W, Benham C, Batchelor E, Levens D. FUBP1 and FUBP2 enforce distinct epigenetic setpoints for MYC expression in primary single murine cells. Commun Biol 2020; 3:545. [PMID: 33005010 PMCID: PMC7530719 DOI: 10.1038/s42003-020-01264-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/01/2020] [Indexed: 11/24/2022] Open
Abstract
Physiologically, MYC levels must be precisely set to faithfully amplify the transcriptome, but in cancer MYC is quantitatively misregulated. Here, we study the variation of MYC amongst single primary cells (B-cells and murine embryonic fibroblasts, MEFs) for the repercussions of variable cellular MYC-levels and setpoints. Because FUBPs have been proposed to be molecular “cruise controls” that constrain MYC expression, their role in determining basal or activated MYC-levels was also examined. Growing cells remember low and high-MYC setpoints through multiple cell divisions and are limited by the same expression ceiling even after modest MYC-activation. High MYC MEFs are enriched for mRNAs regulating inflammation and immunity. After strong stimulation, many cells break through the ceiling and intensify MYC expression. Lacking FUBPs, unstimulated MEFs express levels otherwise attained only with stimulation and sponsor MYC chromatin changes, revealed by chromatin marks. Thus, the FUBPs enforce epigenetic setpoints that restrict MYC expression. Ying Zheng et al. characterize MYC gene and protein expression in single mammalian cells in response to various external signals. They find that individual cells show either high or low basal MYC expression setpoints, and that adherence to these setpoints as well as the magnitude of the response of MYC to stimulation, is controlled by FUBP1 and FUBP2.
Collapse
Affiliation(s)
- Ying Zheng
- Lab of Pathology, National Cancer Institutes, Bethesda, MD, USA
| | - Wendy Dubois
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institutes, Bethesda, MD, USA
| | - Craig Benham
- Biomedical Engineering, University of California, Davis, CA, USA
| | - Eric Batchelor
- Masonic Cancer Center and Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - David Levens
- Lab of Pathology, National Cancer Institutes, Bethesda, MD, USA.
| |
Collapse
|
9
|
Comparative structural analyses and nucleotide-binding characterization of the four KH domains of FUBP1. Sci Rep 2020; 10:13459. [PMID: 32778776 PMCID: PMC7417555 DOI: 10.1038/s41598-020-69832-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
The FUBP1-FUSE complex is an essential component of a transcription molecular machinery that is necessary for tight regulation of expression of many key genes including c-Myc and p21. FUBP1 utilizes its four articulated KH modules, which function cooperatively, for FUSE nucleotide binding. To understand molecular mechanisms fundamental to the intermolecular interaction, we present a set of crystal structures, as well ssDNA-binding characterization of FUBP1 KH domains. All KH1-4 motifs were highly topologically conserved, and were able to interact with FUSE individually and independently. Nevertheless, differences in nucleotide binding properties among the four KH domains were evident, including higher nucleotide-binding potency for KH3 as well as diverse nucleotide sequence preferences. Variations in amino acid compositions at one side of the binding cleft responsible for nucleobase resulted in diverse shapes and electrostatic charge interaction, which might feasibly be a contributing factor for different nucleotide-binding propensities among KH1-4. Nonetheless, conservation of structure and nucleotide-binding property in all four KH motifs is essential for the cooperativity of multi KH modules present in FUBP1 towards nanomolar affinity for FUSE interaction. Comprehensive structural comparison and ssDNA binding characteristics of all four KH domains presented here provide molecular insights at a fundamental level that might be beneficial for elucidating the mechanisms of the FUBP1-FUSE interaction.
Collapse
|
10
|
Swiatkowska A, Dutkiewicz M, Machtel P, Janecki DM, Kabacinska M, Żydowicz-Machtel P, Ciesiołka J. Regulation of the p53 expression profile by hnRNP K under stress conditions. RNA Biol 2020; 17:1402-1415. [PMID: 32449427 DOI: 10.1080/15476286.2020.1771944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The p53 protein is one of the transcription factors responsible for cell cycle regulation and prevention of cancer development. Its expression is regulated at the transcriptional, translational and post-translational levels. Recent years of research have shown that the 5' terminus of p53 mRNA plays an important role in this regulation. This region seems to be a docking platform for proteins involved in p53 expression, particularly under stress conditions. Here, we applied RNA-centric affinity chromatography to search for proteins that bind to the 5' terminus of p53 mRNA and thus may be able to regulate the p53 expression profile. We found heterogeneous nuclear ribonucleoprotein K, hnRNP K, to be one of the top candidates. Binding of hnRNP K to the 5'-terminal region of p53 mRNA was confirmed in vitro. We demonstrated that changes in the hnRNP K level in the cell strongly affected the p53 expression profile under various stress conditions. Downregulation or overexpression of hnRNP K caused a decrease or an increase in the p53 mRNA amount, respectively, pointing to the transcriptional mode of expression regulation. However, when hnRNP K was overexpressed under endoplasmic reticulum stress and the p53 amount has elevated no changes in the p53 mRNA level were detected suggesting translational regulation of p53 expression. Our findings have shown that hnRNP K is not only a mutual partner of p53 in the transcriptional activation of target genes under stress conditions but it also acts as a regulator of p53 expression at the transcriptional and potentially translational levels.
Collapse
Affiliation(s)
- Agata Swiatkowska
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| | - Mariola Dutkiewicz
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| | - Piotr Machtel
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| | - Damian M Janecki
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| | - Martyna Kabacinska
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| | | | - Jerzy Ciesiołka
- Polish Academy of Sciences, Institute of Bioorganic Chemistry , Poznan, Poland
| |
Collapse
|
11
|
Li J, Zhang W, Liu W, Rong J, Chen Y, Gu W, Zhang W. Association of Leukemia Target Genes Tet2, Bcl2, and Slc23a2 in Vitamin C Pathways. Cancer Genomics Proteomics 2019; 16:333-344. [PMID: 31467227 DOI: 10.21873/cgp.20138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Vitamin C has been used in combination with several target genes in the treatment of leukemia. Tet methylcytosine dioxygenase (Tet2), B-cell lymphoma 2 (Bcl2), and solute carrier family 23 member 2 (Slc23a2) are the major target genes in the treatment of leukemia and are relevant to vitamin C. MATERIALS AND METHODS Using whole-genome expression profiles from mouse livers, the expression quantitative trait locus (eQTL), correlation matrix, and gene network graph were constructed with probes from each of these three genes and with their relative genes. The function of key genes was examined by their pathways and reported information. The results indicated that although direct correlations among their expression levels were not strong, alternative connecting pathways were discovered. By comparing the expression levels of one probe with known sequences from each of the three genes, we identified several key genes, induced myeloid leukemia cell differentiation protein (Mcl1), far upstream element-binding protein 1 (Fubp1), and tumor protein D52-like 2 (Tpd52l2), which play important roles in acute lymphocytic leukemia and acute myelocytic leukemia. In conclusion, Alternative pathways and key genes that connect Tet2, Bcl2, and Slc23a2 for their therapeutic applications with vitamin C were identified.
Collapse
Affiliation(s)
- Jing Li
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Wenqi Zhang
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A.,College of Basic Medicine, Hebei Medical University, Shijiazhuang, P.R. China
| | - Weidong Liu
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A.,The First Hospital of Qiqihar City, Heilongjiang, P.R. China
| | - Jarrett Rong
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A.,BSA Biology, Health Science Scholars Honors Program, the University of Texas at Austin, Austin, TX, U.S.A
| | - Yuhan Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, P.R. China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A. .,Research Service, Memphis VA Medical Center, Memphis, TN, U.S.A
| | - Wei Zhang
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, U.S.A. .,Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, P.R. China
| |
Collapse
|
12
|
Schmidt L, Heyes E, Scheiblecker L, Eder T, Volpe G, Frampton J, Nerlov C, Valent P, Grembecka J, Grebien F. CEBPA-mutated leukemia is sensitive to genetic and pharmacological targeting of the MLL1 complex. Leukemia 2019; 33:1608-1619. [PMID: 30679799 PMCID: PMC6612510 DOI: 10.1038/s41375-019-0382-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/28/2018] [Accepted: 12/24/2018] [Indexed: 12/12/2022]
Abstract
The gene encoding the transcription factor C/EBPα is mutated in 10-15% of acute myeloid leukemia (AML) patients. N-terminal CEBPA mutations cause ablation of full-length C/EBPα without affecting the expression of a shorter oncogenic isoform, termed p30. The mechanistic basis of p30-induced leukemogenesis is incompletely understood. Here, we demonstrate that the MLL1 histone-methyltransferase complex represents a critical actionable vulnerability in CEBPA-mutated AML. Oncogenic C/EBPα p30 and MLL1 show global co-localization on chromatin and p30 exhibits robust physical interaction with the MLL1 complex. CRISPR/Cas9-mediated mutagenesis of MLL1 results in proliferation arrest and myeloid differentiation in C/EBPα p30-expressing cells. In line, CEBPA-mutated hematopoietic progenitor cells are hypersensitive to pharmacological targeting of the MLL1 complex. Inhibitor treatment impairs proliferation and restores myeloid differentiation potential in mouse and human AML cells with CEBPA mutations. Finally, we identify the transcription factor GATA2 as a direct critical target of the p30-MLL1 interaction. Altogether, we show that C/EBPα p30 requires the MLL1 complex to regulate oncogenic gene expression and that CEBPA-mutated AML is hypersensitive to perturbation of the MLL1 complex. These findings identify the MLL1 complex as a potential therapeutic target in AML with CEBPA mutations.
Collapse
Affiliation(s)
- Luisa Schmidt
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, Austria
| | - Elizabeth Heyes
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | | | - Thomas Eder
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Giacomo Volpe
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, Birmingham, UK
- Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences and Guangzhou Medical University, Guangzhou, China
| | - Jon Frampton
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, Birmingham, UK
| | - Claus Nerlov
- Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.
- Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
13
|
Apobec3A maintains HIV-1 latency through recruitment of epigenetic silencing machinery to the long terminal repeat. Proc Natl Acad Sci U S A 2019; 116:2282-2289. [PMID: 30670656 DOI: 10.1073/pnas.1819386116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
HIV-1 integrates into the genome of target cells and establishes latency indefinitely. Understanding the molecular mechanism of HIV-1 latency maintenance is needed for therapeutic strategies to combat existing infection. In this study, we found an unexpected role for Apobec3A (apolipoprotein B MRNA editing enzyme catalytic subunit 3A, abbreviated "A3A") in maintaining the latency state within HIV-1-infected cells. Overexpression of A3A in latently infected cell lines led to lower reactivation, while knockdown or knockout of A3A led to increased spontaneous and inducible HIV-1 reactivation. A3A maintains HIV-1 latency by associating with proviral DNA at the 5' long terminal repeat region, recruiting KAP1 and HP1, and imposing repressive histone marks. We show that knockdown of A3A in latently infected human primary CD4 T cells enhanced HIV-1 reactivation. Collectively, we provide evidence and a mechanism by which A3A reinforces HIV-1 latency in infected CD4 T cells.
Collapse
|
14
|
Steiner M, Schneider L, Yillah J, Gerlach K, Kuvardina ON, Meyer A, Maring A, Bonig H, Seifried E, Zörnig M, Lausen J. FUSE binding protein 1 (FUBP1) expression is upregulated by T-cell acute lymphocytic leukemia protein 1 (TAL1) and required for efficient erythroid differentiation. PLoS One 2019; 14:e0210515. [PMID: 30653565 PMCID: PMC6336336 DOI: 10.1371/journal.pone.0210515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/23/2018] [Indexed: 11/18/2022] Open
Abstract
During erythropoiesis, haematopoietic stem cells (HSCs) differentiate in successive steps of commitment and specification to mature erythrocytes. This differentiation process is controlled by transcription factors that establish stage- and cell type-specific gene expression. In this study, we demonstrate that FUSE binding protein 1 (FUBP1), a transcriptional regulator important for HSC self-renewal and survival, is regulated by T-cell acute lymphocytic leukaemia 1 (TAL1) in erythroid progenitor cells. TAL1 directly activates the FUBP1 promoter, leading to increased FUBP1 expression during erythroid differentiation. The binding of TAL1 to the FUBP1 promoter is highly dependent on an intact GATA sequence in a combined E-box/GATA motif. We found that FUBP1 expression is required for efficient erythropoiesis, as FUBP1-deficient progenitor cells were limited in their potential of erythroid differentiation. Thus, the finding of an interconnection between GATA1/TAL1 and FUBP1 reveals a molecular mechanism that is part of the switch from progenitor- to erythrocyte-specific gene expression. In summary, we identified a TAL1/FUBP1 transcriptional relationship, whose physiological function in haematopoiesis is connected to proper erythropoiesis.
Collapse
Affiliation(s)
- Marlene Steiner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Lucas Schneider
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Jasmin Yillah
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Katharina Gerlach
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Olga N. Kuvardina
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Annekarin Meyer
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Alisa Maring
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Erhard Seifried
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
- * E-mail: (MZ); (JL)
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
- * E-mail: (MZ); (JL)
| |
Collapse
|
15
|
Hoang VT, Verma D, Godavarthy PS, Llavona P, Steiner M, Gerlach K, Michels BE, Bohnenberger H, Wachter A, Oellerich T, Müller-Kuller U, Weissenberger E, Voutsinas JM, Oehler VG, Farin HF, Zörnig M, Krause DS. The transcriptional regulator FUBP1 influences disease outcome in murine and human myeloid leukemia. Leukemia 2019; 33:1700-1712. [PMID: 30635626 DOI: 10.1038/s41375-018-0358-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/28/2018] [Accepted: 12/05/2018] [Indexed: 01/20/2023]
Abstract
The transcriptional regulator far upstream element binding protein 1 (FUBP1) acts as an oncoprotein in solid tumor entities and plays a role in the maintenance of hematopoietic stem cells. However, its potential function in leukemia is unknown. In murine models of chronic (CML) and acute myeloid leukemia (AML) induced by BCR-ABL1 and MLL-AF9, respectively, knockdown of Fubp1 resulted in prolonged survival, decreased numbers of CML progenitor cells, decreased cell cycle activity and increased apoptosis. Knockdown of FUBP1 in CML and AML cell lines recapitulated these findings and revealed enhanced DNA damage compared to leukemia cells expressing wild type FUBP1 levels. FUBP1 was more highly expressed in human CML compared to normal bone marrow cells and its expression correlated with disease progression. In AML, higher FUBP1 expression in patient leukemia cells was observed with a trend toward correlation with shorter overall survival. Treatment of mice with AML with irinotecan, known to inhibit topoisomerase I and FUBP1, significantly prolonged survival alone or in combination with cytarabine. In summary, our data suggest that FUBP1 acts as cell cycle regulator and apoptosis inhibitor in leukemia. We demonstrated that FUBP1 might play a role in DNA repair, and its inhibition may improve outcome in leukemia patients.
Collapse
Affiliation(s)
- Van T Hoang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Divij Verma
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | | | - Pablo Llavona
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Marlene Steiner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Katharina Gerlach
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Birgitta E Michels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Hanibal Bohnenberger
- Universitätsmedizin Göttingen, Institute of Pathology, Georg-August-Universität, 37075, Göttingen, Germany
| | - Astrid Wachter
- Universitätsmedizin Göttingen, Department of Medical Statistics, Georg-August-Universität, 37075, Göttingen, Germany
| | - Thomas Oellerich
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Hospital Frankfurt, Department of Medicine II, Hematology/Oncology, Frankfurt, Germany
| | - Uta Müller-Kuller
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Eva Weissenberger
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Jenna M Voutsinas
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Biostatistics, Seattle, WA, USA
| | - Vivian G Oehler
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Division of Hematology, University of Washington Medical Center, Seattle, WA, USA
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt, Germany.
| |
Collapse
|
16
|
Debaize L, Troadec MB. The master regulator FUBP1: its emerging role in normal cell function and malignant development. Cell Mol Life Sci 2019; 76:259-281. [PMID: 30343319 PMCID: PMC11105487 DOI: 10.1007/s00018-018-2933-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/06/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022]
Abstract
The human Far Upstream Element (FUSE) Binding Protein 1 (FUBP1) is a multifunctional DNA- and RNA-binding protein involved in diverse cellular processes. FUBP1 is a master regulator of transcription, translation, and RNA splicing. FUBP1 has been identified as a potent pro-proliferative and anti-apoptotic factor by modulation of complex networks. FUBP1 is also described either as an oncoprotein or a tumor suppressor. Especially, FUBP1 overexpression is observed in a growing number of cancer and leads to a deregulation of targets that includes the fine-tuned MYC oncogene. Moreover, recent loss-of-function analyses of FUBP1 establish its essential functions in hematopoietic stem cell maintenance and survival. Therefore, FUBP1 appears as an emerging suspect in hematologic disorders in addition to solid tumors. The scope of the present review is to describe the advances in our understanding of the molecular basis of FUBP1 functions in normal cells and carcinogenesis. We also delineate the recent progresses in the understanding of the master role of FUBP1 in normal and pathological hematopoiesis. We conclude that FUBP1 is not only worth studying biologically but is also of clinical relevance through its pivotal role in regulating multiple cellular processes and its involvement in oncogenesis.
Collapse
Affiliation(s)
- Lydie Debaize
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, F-35000, Rennes, France
| | - Marie-Bérengère Troadec
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, F-35000, Rennes, France.
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France.
- CHRU de Brest, laboratoire de cytogénétique, F-29200, Brest, France.
| |
Collapse
|
17
|
Debaize L, Jakobczyk H, Avner S, Gaudichon J, Rio AG, Sérandour AA, Dorsheimer L, Chalmel F, Carroll JS, Zörnig M, Rieger MA, Delalande O, Salbert G, Galibert MD, Gandemer V, Troadec MB. Interplay between transcription regulators RUNX1 and FUBP1 activates an enhancer of the oncogene c-KIT and amplifies cell proliferation. Nucleic Acids Res 2018; 46:11214-11228. [PMID: 30500954 PMCID: PMC6265458 DOI: 10.1093/nar/gky756] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/31/2022] Open
Abstract
Runt-related transcription factor 1 (RUNX1) is a well-known master regulator of hematopoietic lineages but its mechanisms of action are still not fully understood. Here, we found that RUNX1 localizes on active chromatin together with Far Upstream Binding Protein 1 (FUBP1) in human B-cell precursor lymphoblasts, and that both factors interact in the same transcriptional regulatory complex. RUNX1 and FUBP1 chromatin localization identified c-KIT as a common target gene. We characterized two regulatory regions, at +700 bp and +30 kb within the first intron of c-KIT, bound by both RUNX1 and FUBP1, and that present active histone marks. Based on these regions, we proposed a novel FUBP1 FUSE-like DNA-binding sequence on the +30 kb enhancer. We demonstrated that FUBP1 and RUNX1 cooperate for the regulation of the expression of the oncogene c-KIT. Notably, upregulation of c-KIT expression by FUBP1 and RUNX1 promotes cell proliferation and renders cells more resistant to the c-KIT inhibitor imatinib mesylate, a common therapeutic drug. These results reveal a new mechanism of action of RUNX1 that implicates FUBP1, as a facilitator, to trigger transcriptional regulation of c-KIT and to regulate cell proliferation. Deregulation of this regulatory mechanism may explain some oncogenic function of RUNX1 and FUBP1.
Collapse
Affiliation(s)
- Lydie Debaize
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Hélène Jakobczyk
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Stéphane Avner
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Jérémie Gaudichon
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Anne-Gaëlle Rio
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Aurélien A Sérandour
- CRCINA, INSERM, CNRS, Université d’Angers, Université de Nantes, 44035 Nantes, France
- Ecole Centrale de Nantes, Nantes, France
| | - Lena Dorsheimer
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085, F-35000 Rennes, France
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, D-60528 Frankfurt, Germany
| | - Michael A Rieger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Olivier Delalande
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Gilles Salbert
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| | - Marie-Dominique Galibert
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
- Génétique Somatique des Cancers, Centre Hospitalier Universitaire, 35033 Rennes, France
| | - Virginie Gandemer
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
- Department of pediatric oncohematology, Centre Hospitalier Universitaire, 35203 Rennes, France
| | - Marie-Bérengère Troadec
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) – UMR 6290, F-35000 Rennes, France
| |
Collapse
|
18
|
Hwang I, Cao D, Na Y, Kim DY, Zhang T, Yao J, Oh H, Hu J, Zheng H, Yao Y, Paik J. Far Upstream Element-Binding Protein 1 Regulates LSD1 Alternative Splicing to Promote Terminal Differentiation of Neural Progenitors. Stem Cell Reports 2018; 10:1208-1221. [PMID: 29606613 PMCID: PMC5998560 DOI: 10.1016/j.stemcr.2018.02.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/08/2023] Open
Abstract
Loss of a cell's ability to terminally differentiate because of mutations is a selected genetic event in tumorigenesis. Genomic analyses of low-grade glioma have reported recurrent mutations of far upstream element-binding protein 1 (FUBP1). Here, we show that FUBP1 expression is dynamically regulated during neurogenesis and that its downregulation in neural progenitors impairs terminal differentiation and promotes tumorigenesis collaboratively with expression of IDH1R132H. Mechanistically, collaborative action between SRRM4 and FUBP1 is necessary for mini-exon splicing of the neurospecific LSD1+8a isoform. LSD1+8a was downregulated upon loss of FUBP1 in neural progenitors, thereby impairing terminal neuronal differentiation and maturation. Reinforcing LSD1+8a expression in FUBP1-downregulated neural progenitors restored terminal differentiation and suppressed tumorigenesis; hence, LSD1+8a is an obligatory effector of FUBP1-dependent neuronal differentiation. These findings establish a direct role for FUBP1 in neuronal differentiation and also explain its tumor-suppressor function in the nervous system.
Collapse
Affiliation(s)
- Inah Hwang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dongqing Cao
- Neurosurgical Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Yoonmi Na
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hwanhee Oh
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jian Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hongwu Zheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Yu Yao
- Neurosurgical Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
19
|
Putzbach W, Gao QQ, Patel M, van Dongen S, Haluck-Kangas A, Sarshad AA, Bartom ET, Kim KYA, Scholtens DM, Hafner M, Zhao JC, Murmann AE, Peter ME. Many si/shRNAs can kill cancer cells by targeting multiple survival genes through an off-target mechanism. eLife 2017; 6. [PMID: 29063830 PMCID: PMC5655136 DOI: 10.7554/elife.29702] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 09/13/2017] [Indexed: 12/27/2022] Open
Abstract
Over 80% of multiple-tested siRNAs and shRNAs targeting CD95 or CD95 ligand (CD95L) induce a form of cell death characterized by simultaneous activation of multiple cell death pathways preferentially killing transformed and cancer stem cells. We now show these si/shRNAs kill cancer cells through canonical RNAi by targeting the 3’UTR of critical survival genes in a unique form of off-target effect we call DISE (death induced by survival gene elimination). Drosha and Dicer-deficient cells, devoid of most miRNAs, are hypersensitive to DISE, suggesting cellular miRNAs protect cells from this form of cell death. By testing 4666 shRNAs derived from the CD95 and CD95L mRNA sequences and an unrelated control gene, Venus, we have identified many toxic sequences - most of them located in the open reading frame of CD95L. We propose that specific toxic RNAi-active sequences present in the genome can kill cancer cells. Cells store their genetic code within molecules of DNA. Some of this information will be copied into chemically similar molecules called RNAs, from which the sequence of letters in the genetic code can be translated to build proteins. However, these messenger RNAs are not the only RNA molecules that cells can make. MicroRNAs are other short pieces of RNA that closely match sequences in parts of certain messenger RNAs. The messenger RNAs targeted by microRNAs are broken down inside the cell, which reduces how much protein can be produced from them. Since its discovery, scientists have exploited this process – called RNA interference (or RNAi for short) – and designed microRNA-like small interfering RNAs (siRNAs) to target particular messenger RNAs and decrease the levels of the corresponding proteins in countless experiments. Two proteins that have been studied in RNAi experiments are CD95 and its interaction partner CD95L. Both of these proteins are important in human cancer cells, and targeting them via RNAi killed cancer cells in an unknown mechanism that the cancer cells were unable to resist. RNAi experiments are designed to be specific, but sometimes they can accidently target other non-target messenger RNAs. Putzbach, Gao, Patel et al. have now analyzed all of the siRNAs that can be made from the messenger RNAs for CD95 and CD95L to mediate RNAi in cancer cells. This revealed that several messenger RNAs, other than those for CD95 and CD95L, were unintentionally being targeted, including many that code for proteins that cells need to survive. Further examination of the messenger RNA for CD95 and CD95L showed that they contain short sequences that are similar to those in the messenger RNAs of the genes that encode these survival proteins. Putzbach et al. were able to study and then predict which siRNA sequences would be toxic to cancer cells. These findings indicate that an RNAi off-target effect may actually be used to kill cancer cells. Future studies will determine whether this effect could be exploited to shrink tumors in animal models of cancer. If successful, this in turn could lead to new treatments for cancer patients.
Collapse
Affiliation(s)
- William Putzbach
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Quan Q Gao
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Monal Patel
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Stijn van Dongen
- European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Ashley Haluck-Kangas
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Aishe A Sarshad
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, United States
| | - Kwang-Youn A Kim
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Denise M Scholtens
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Markus Hafner
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Jonathan C Zhao
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Andrea E Murmann
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States
| | - Marcus E Peter
- Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, United States.,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, United States
| |
Collapse
|
20
|
Khageh Hosseini S, Kolterer S, Steiner M, von Manstein V, Gerlach K, Trojan J, Waidmann O, Zeuzem S, Schulze JO, Hahn S, Steinhilber D, Gatterdam V, Tampé R, Biondi RM, Proschak E, Zörnig M. Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Biochem Pharmacol 2017; 146:53-62. [PMID: 29031818 DOI: 10.1016/j.bcp.2017.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/11/2017] [Indexed: 01/03/2023]
Abstract
The transcriptional regulator FUSE Binding Protein 1 (FUBP1) is overexpressed in more than 80% of all human hepatocellular carcinomas (HCCs) and other solid tumor entities including prostate and colorectal carcinoma. FUBP1 expression is required for HCC tumor cell expansion, and it functions as an important pro-proliferative and anti-apoptotic oncoprotein that binds to the single-stranded DNA sequence FUSE to regulate the transcription of a variety of target genes. In this study, we screened an FDA-approved drug library and discovered that the Topoisomerase I (TOP1) inhibitor camptothecin (CPT) and its derivative 7-ethyl-10-hydroxycamptothecin (SN-38), the active irinotecan metabolite that is used in the clinics in combination with other chemotherapeutics to treat carcinoma, inhibit FUBP1 activity. Both molecules prevent in vitro the binding of FUBP1 to its single-stranded target DNA FUSE, and they induce deregulation of FUBP1 target genes in HCC cells. Our results suggest the interference with the FUBP1/FUSE interaction as a further molecular mechanism that, in addition to the inactivation of TOP1, may contribute to the therapeutic potential of CPT/SN-38. Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression.
Collapse
Affiliation(s)
- Sabrina Khageh Hosseini
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Stefanie Kolterer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Marlene Steiner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Viktoria von Manstein
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Katharina Gerlach
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Jörg Trojan
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Oliver Waidmann
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Jörg O Schulze
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Steffen Hahn
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Volker Gatterdam
- Institute of Biochemistry, Biocenter/Cluster of Excellence-Macromolecular Complexes, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter/Cluster of Excellence-Macromolecular Complexes, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Ricardo M Biondi
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
21
|
Zhang J, Xiong X, Hua X, Cao W, Qin S, Dai L, Liang P, Zhang H, Liu Z. Knockdown of FUSE binding protein 1 enhances the sensitivity of epithelial ovarian cancer cells to carboplatin. Oncol Lett 2017; 14:5819-5824. [PMID: 29113212 PMCID: PMC5661610 DOI: 10.3892/ol.2017.6978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) affects almost 25,000 women annually and is the fifth most common malignancy in women in North America. A combination of surgery and cytotoxic chemotherapy may produce a favorable clinical response. The platinum-paclitaxel combination regimen is the chemotherapy gold-standard for advanced ovarian cancer, and carboplatin is one of the agents in this combination therapy. However, the majority of patients eventually experience a relapse due to the development of platinum resistance. FUSE binding protein 1 (FBP1) has been identified as an anti-apoptotic and pro-proliferative oncoprotein that is overexpressed in hepatocellular carcinoma. Its high expression is also associated with carboplatin resistance. In the present study, it was identified that the expression of FBP1 was significantly higher in EOC tissues than in normal epithelial ovarian or in epithelial ovarian adenoma tissue. FBP1 expression was significantly correlated with the grade of epithelial ovarian cancer. Carboplatin inhibited the expression of FBP1 in epithelial ovarian cancer cells and the knockdown of FBP1 enhanced the inhibition of cell viability and migration by carboplatin. In addition to FBP1, carboplatin also inhibited the expression of β-catenin and matrix metalloproteinase (MMP)-9. Furthermore, the expression of β-catenin and MMP-9 were lower in FBP1 knockdown cells compared with control EOC cells. FBP1 may thus serve a role in the regulation of the expression of β-catenin and MMP-9; the inhibition of β-catenin and MMP-9 by carboplatin may be mediated through the inhibition of FBP1. The inhibition of FBP1 expression by carboplatin may be a mechanism in the treatment of EOC by carboplatin.
Collapse
Affiliation(s)
- Jinli Zhang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Xing Hua
- Department of Pathology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Wenjuan Cao
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Shengnan Qin
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Libing Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Peihong Liang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Huiling Zhang
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| |
Collapse
|
22
|
Lunger I, Fawaz M, Rieger MA. Single-cell analyses to reveal hematopoietic stem cell fate decisions. FEBS Lett 2017; 591:2195-2212. [PMID: 28600837 DOI: 10.1002/1873-3468.12712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/19/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are the best studied adult stem cells with enormous clinical value. Most of our knowledge about their biology relies on assays at the single HSC level. However, only the recent advances in developing new single cell technologies allowed the elucidation of the complex regulation of HSC fate decision control. This Review will focus on current attempts to investigate individual HSCs at molecular and functional levels. The advantages of these technologies leading to groundbreaking insights into hematopoiesis will be highlighted, and the challenges facing these technologies will be discussed. The importance of combining molecular and functional assays to enlighten regulatory networks of HSC fate decision control, ideally at high temporal resolution, becomes apparent for future studies.
Collapse
Affiliation(s)
- Ilaria Lunger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Malak Fawaz
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael A Rieger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Delayed Mesoderm and Erythroid Differentiation of Murine Embryonic Stem Cells in the Absence of the Transcriptional Regulator FUBP1. Stem Cells Int 2017; 2017:5762301. [PMID: 28588622 PMCID: PMC5447289 DOI: 10.1155/2017/5762301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/02/2017] [Accepted: 03/19/2017] [Indexed: 11/18/2022] Open
Abstract
The transcriptional regulator far upstream binding protein 1 (FUBP1) is essential for fetal and adult hematopoietic stem cell (HSC) self-renewal, and the constitutive absence of FUBP1 activity during early development leads to embryonic lethality in homozygous mutant mice. To investigate the role of FUBP1 in murine embryonic stem cells (ESCs) and in particular during differentiation into hematopoietic lineages, we generated Fubp1 knockout (KO) ESC clones using CRISPR/Cas9 technology. Although FUBP1 is expressed in undifferentiated ESCs and during spontaneous differentiation following aggregation into embryoid bodies (EBs), absence of FUBP1 did not affect ESC maintenance. Interestingly, we observed a delayed differentiation of FUBP1-deficient ESCs into the mesoderm germ layer, as indicated by impaired expression of several mesoderm markers including Brachyury at an early time point of ESC differentiation upon aggregation to EBs. Coculture experiments with OP9 cells in the presence of erythropoietin revealed a diminished differentiation capacity of Fubp1 KO ESCs into the erythroid lineage. Our data showed that FUBP1 is important for the onset of mesoderm differentiation and maturation of hematopoietic progenitor cells into the erythroid lineage, a finding that is supported by the phenotype of FUBP1-deficient mice.
Collapse
|
24
|
Zaytseva O, Quinn LM. Controlling the Master: Chromatin Dynamics at the MYC Promoter Integrate Developmental Signaling. Genes (Basel) 2017; 8:genes8040118. [PMID: 28398229 PMCID: PMC5406865 DOI: 10.3390/genes8040118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/15/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023] Open
Abstract
The transcription factor and cell growth regulator MYC is potently oncogenic and estimated to contribute to most cancers. Decades of attempts to therapeutically target MYC directly have not resulted in feasible clinical applications, and efforts have moved toward indirectly targeting MYC expression, function and/or activity to treat MYC-driven cancer. A multitude of developmental and growth signaling pathways converge on the MYC promoter to modulate transcription through their downstream effectors. Critically, even small increases in MYC abundance (<2 fold) are sufficient to drive overproliferation; however, the details of how oncogenic/growth signaling networks regulate MYC at the level of transcription remain nebulous even during normal development. It is therefore essential to first decipher mechanisms of growth signal-stimulated MYC transcription using in vivo models, with intact signaling environments, to determine exactly how these networks are dysregulated in human cancer. This in turn will provide new modalities and approaches to treat MYC-driven malignancy. Drosophila genetic studies have shed much light on how complex networks signal to transcription factors and enhancers to orchestrate Drosophila MYC (dMYC) transcription, and thus growth and patterning of complex multicellular tissue and organs. This review will discuss the many pathways implicated in patterning MYC transcription during development and the molecular events at the MYC promoter that link signaling to expression. Attention will also be drawn to parallels between mammalian and fly regulation of MYC at the level of transcription.
Collapse
Affiliation(s)
- Olga Zaytseva
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| | - Leonie M Quinn
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
25
|
Abstract
Drosophila genetic studies demonstrate that cell and tissue growth regulation is a primary developmental function of P-element somatic inhibitor (Psi), the sole ortholog of FUBP family RNA/DNA-binding proteins. Psi achieves growth control through interaction with Mediator, observations that should put to rest controversy surrounding Pol II transcriptional functions for these KH domain proteins.
Collapse
Affiliation(s)
- Leonie M Quinn
- a Department of Cancer Biology and Therapeutics , The John Curtin School of Medical Research, The Australian National University , Canberra , ACT , Australia
| |
Collapse
|
26
|
Pyrazolo[1,5 a ]pyrimidines as a new class of FUSE binding protein 1 (FUBP1) inhibitors. Bioorg Med Chem 2016; 24:5717-5729. [DOI: 10.1016/j.bmc.2016.09.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/13/2016] [Accepted: 09/08/2016] [Indexed: 12/12/2022]
|
27
|
Wang J, Li X, Wang L, Li J, Zhao Y, Bou G, Li Y, Jiao G, Shen X, Wei R, Liu S, Xie B, Lei L, Li W, Zhou Q, Liu Z. A novel long intergenic noncoding RNA indispensable for the cleavage of mouse two-cell embryos. EMBO Rep 2016; 17:1452-1470. [PMID: 27496889 DOI: 10.15252/embr.201642051] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/07/2016] [Indexed: 01/30/2023] Open
Abstract
Endogenous retroviruses (ERVs) are transcriptionally active in cleavage stage embryos, yet their functions are unknown. ERV sequences are present in the majority of long intergenic noncoding RNAs (lincRNAs) in mouse and humans, playing key roles in many cellular processes and diseases. Here, we identify LincGET as a nuclear lincRNA that is GLN-, MERVL-, and ERVK-associated and essential for mouse embryonic development beyond the two-cell stage. LincGET is expressed in late two- to four-cell mouse embryos. Its depletion leads to developmental arrest at the late G2 phase of the two-cell stage and to MAPK signaling pathway inhibition. LincGET forms an RNA-protein complex with hnRNP U, FUBP1, and ILF2, promoting the cis-regulatory activity of long terminal repeats (LTRs) in GLN, MERVL, and ERVK (GLKLTRs), and inhibiting RNA alternative splicing, partially by downregulating hnRNP U, FUBP1, and ILF2 protein levels. Hnrnpu or Ilf2 mRNA injection at the pronuclear stage also decreases the preimplantation developmental rate, and Fubp1 mRNA injection at the pronuclear stage causes a block at the two-cell stage. Thus, as the first functional ERV-associated lincRNA, LincGET provides clues for ERV functions in cleavage stage embryonic development.
Collapse
Affiliation(s)
- Jiaqiang Wang
- College of Life Science, Northeast Agricultural University, Harbin, China State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xin Li
- College of Life Science, Northeast Agricultural University, Harbin, China State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Leyun Wang
- College of Life Science, Northeast Agricultural University, Harbin, China State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jingyu Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yanhua Zhao
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Gerelchimeg Bou
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yufei Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guanyi Jiao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinghui Shen
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Renyue Wei
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Shichao Liu
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Bingteng Xie
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Lei Lei
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Wei Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
28
|
|
29
|
Zhou W, Chung YJ, Parrilla Castellar ER, Zheng Y, Chung HJ, Bandle R, Liu J, Tessarollo L, Batchelor E, Aplan PD, Levens D. Far Upstream Element Binding Protein Plays a Crucial Role in Embryonic Development, Hematopoiesis, and Stabilizing Myc Expression Levels. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:701-15. [PMID: 26774856 DOI: 10.1016/j.ajpath.2015.10.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/29/2015] [Accepted: 10/27/2015] [Indexed: 11/27/2022]
Abstract
The transcription factor far upstream element binding protein (FBP) binds and activates the MYC promoter when far upstream element is via TFIIH helicase activity early in the transcription cycle. The fundamental biology and pathology of FBP are complex. In some tumors FBP seems pro-oncogenic, whereas in others it is a tumor suppressor. We generated an FBP knockout (Fubp1(-/-)) mouse to study FBP deficiency. FBP is embryo lethal from embryonic day 10.5 to birth. A spectrum of pathology is associated with FBP loss; besides cerebral hyperplasia and pulmonary hypoplasia, pale livers, hypoplastic spleen, thymus, and bone marrow, cardiac hypertrophy, placental distress, and small size were all indicative of anemia. Immunophenotyping of hematopoietic cells in wild-type versus knockout livers revealed irregular trilineage anemia, with deficits in colony formation. Despite normal numbers of hematopoietic stem cells, transplantation of Fubp1(-/-) hematopoietic stem cells into irradiated mice entirely failed to reconstitute hematopoiesis. In competitive transplantation assays against wild-type donor bone marrow, Fubp1(-/-) hematopoietic stem cells functioned only sporadically at a low level. Although cultures of wild-type mouse embryo fibroblasts set Myc levels precisely, Myc levels of mouse varied wildly between fibroblasts harvested from different Fubp1(-/-) embryos, suggesting that FBP contributes to Myc set point fixation. FBP helps to hold multiple physiologic processes to close tolerances, at least in part by constraining Myc expression.
Collapse
Affiliation(s)
- Weixin Zhou
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Yang Jo Chung
- Laboratory of Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | - Ying Zheng
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Hye-Jung Chung
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Russell Bandle
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Juhong Liu
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Lino Tessarollo
- Mouse Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Eric Batchelor
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Peter D Aplan
- Laboratory of Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - David Levens
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
30
|
Yamashita M, Nitta E, Suda T. Regulation of hematopoietic stem cell integrity through p53 and its related factors. Ann N Y Acad Sci 2015; 1370:45-54. [DOI: 10.1111/nyas.12986] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/11/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Masayuki Yamashita
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Division of Hematology/Oncology; Department of Medicine, University of California San Francisco; San Francisco California
| | - Eriko Nitta
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- Department of Cellular and Molecular Medicine, Graduate School of Medicine; Chiba University; Chiba Japan
| | - Toshio Suda
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- Cancer Science Institute; National University of Singapore; Singapore
- International Research Center for Medical Sciences; Kumamoto University; Kumamoto Japan
| |
Collapse
|