1
|
Zerella JR, Homan CC, Arts P, Lin X, Spinelli SJ, Venugopal P, Babic M, Brautigan PJ, Truong L, Arriola-Martinez L, Moore S, Hollins R, Parker WT, Nguyen H, Kassahn KS, Branford S, Feurstein S, Larcher L, Sicre de Fontbrune F, Demirdas S, de Munnik S, Antoine-Poirel H, Brichard B, Mansour S, Gordon K, Wlodarski MW, Koppayi A, Dobbins S, Mutsaers PGNJ, Nichols KE, Oak N, DeMille D, Mao R, Crawford A, McCarrier J, Basel D, Flores-Daboub J, Drazer MW, Phillips K, Poplawski NK, Birdsey GM, Pirri D, Ostergaard P, Simons A, Godley LA, Ross DM, Hiwase DK, Soulier J, Brown AL, Carmichael CL, Scott HS, Hahn CN. Germ line ERG haploinsufficiency defines a new syndrome with cytopenia and hematological malignancy predisposition. Blood 2024; 144:1765-1780. [PMID: 38991192 PMCID: PMC11530364 DOI: 10.1182/blood.2024024607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
ABSTRACT The genomics era has facilitated the discovery of new genes that predispose individuals to bone marrow failure (BMF) and hematological malignancy (HM). We report the discovery of ETS-related gene (ERG), a novel, autosomal dominant BMF/HM predisposition gene. ERG is a highly constrained transcription factor that is critical for definitive hematopoiesis, stem cell function, and platelet maintenance. ERG colocalizes with other transcription factors, including RUNX family transcription factor 1 (RUNX1) and GATA binding protein 2 (GATA2), on promoters or enhancers of genes that orchestrate hematopoiesis. We identified a rare heterozygous ERG missense variant in 3 individuals with thrombocytopenia from 1 family and 14 additional ERG variants in unrelated individuals with BMF/HM, including 2 de novo cases and 3 truncating variants. Phenotypes associated with pathogenic germ line ERG variants included cytopenias (thrombocytopenia, neutropenia, and pancytopenia) and HMs (acute myeloid leukemia, myelodysplastic syndrome, and acute lymphoblastic leukemia) with onset before 40 years. Twenty ERG variants (19 missense and 1 truncating), including 3 missense population variants, were functionally characterized. Thirteen potentially pathogenic erythroblast transformation specific (ETS) domain missense variants displayed loss-of-function (LOF) characteristics, thereby disrupting transcriptional transactivation, DNA binding, and/or nuclear localization. Selected variants overexpressed in mouse fetal liver cells failed to drive myeloid differentiation and cytokine-independent growth in culture and to promote acute erythroleukemia when transplanted into mice, concordant with these being LOF variants. Four individuals displayed somatic genetic rescue by copy neutral loss of heterozygosity. Identification of predisposing germ line ERG variants has clinical implications for patient and family diagnoses, counseling, surveillance, and treatment strategies, including selection of bone marrow donors and cell or gene therapy.
Collapse
Affiliation(s)
- Jiarna R. Zerella
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Claire C. Homan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Peer Arts
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Xuzhu Lin
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sam J. Spinelli
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Parvathy Venugopal
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Milena Babic
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Peter J. Brautigan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Lynda Truong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Luis Arriola-Martinez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Sarah Moore
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Rachel Hollins
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Wendy T. Parker
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Hung Nguyen
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Karin S. Kassahn
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Susan Branford
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Simone Feurstein
- Department of Internal Medicine, Section of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lise Larcher
- Université Paris Cité, INSERM and Hôpital Saint-Louis, Assistance Publique–Hôpitaux de Paris, Paris, France
| | | | - Serwet Demirdas
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sonja de Munnik
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Benedicte Brichard
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sahar Mansour
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
- South West Thames Regional Centre for Genomics, St. George's Universities National Health Service Foundation Trust, London, United Kingdom
| | - Kristiana Gordon
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
- Dermatology and Lymphovascular Medicine, St. George's Universities National Health Service Foundation Trust, London, United Kingdom
| | - Marcin W. Wlodarski
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Ashwin Koppayi
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Sara Dobbins
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
| | - Pim G. N. J. Mutsaers
- Department of Hematology, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Kim E. Nichols
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ninad Oak
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Desiree DeMille
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Associated Regional and University Pathologists Laboratories, Salt Lake City, UT
| | - Rong Mao
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Associated Regional and University Pathologists Laboratories, Salt Lake City, UT
- Department of Pathology, University of Utah, Salt Lake City, UT
| | | | - Julie McCarrier
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Donald Basel
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | | | - Michael W. Drazer
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL
| | - Kerry Phillips
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | - Graeme M. Birdsey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Daniela Pirri
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pia Ostergaard
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
| | - Annet Simons
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lucy A. Godley
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - David M. Ross
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Devendra K. Hiwase
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Jean Soulier
- Université Paris Cité, INSERM and Hôpital Saint-Louis, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Anna L. Brown
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Catherine L. Carmichael
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Hamish S. Scott
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Australian Cancer Research Foundation Genomics Facility, Centre for Cancer Biology, Adelaide, SA, Australia
| | - Christopher N. Hahn
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
2
|
León-Gutiérrez G, Elste JE, Cabello-Gutiérrez C, Millán-Pacheco C, Martínez-Gómez MH, Mejía-Alvarez R, Tiwari V, Mejía A. A potent virucidal activity of functionalized TiO 2 nanoparticles adsorbed with flavonoids against SARS-CoV-2. Appl Microbiol Biotechnol 2022; 106:5987-6002. [PMID: 35951081 PMCID: PMC9366830 DOI: 10.1007/s00253-022-12112-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022]
Abstract
Abstract The coronavirus SARS-CoV-2 has caused a pandemic with > 550 millions of cases and > 6 millions of deaths worldwide. Medical management of COVID-19 relies on supportive care as no specific targeted therapies are available yet. Given its devastating effects on the economy and mental health, it is imperative to develop novel antivirals. An ideal candidate will be an agent that blocks the early events of viral attachment and cell entry, thereby preventing viral infection and spread. This work reports functionalized titanium dioxide (TiO2)-based nanoparticles adsorbed with flavonoids that block SARS-CoV-2 entry and fusion. Using molecular docking analysis, two flavonoids were chosen for their specific binding to critical regions of the SARS-CoV-2 spike glycoprotein that interacts with the host cell angiotensin-converting enzyme-2 (ACE-2) receptor. These flavonoids were adsorbed onto TiO2 functionalized nanoparticles (FTNP). This new nanoparticulate compound was assayed in vitro against two different coronaviruses; HCoV 229E and SARS-CoV-2, in both cases a clear antiviral effect was observed. Furthermore, using a reporter-based cell culture model, a potent antiviral activity is demonstrated. The adsorption of flavonoids to functionalized TiO2 nanoparticles induces a ~ threefold increase of that activity. These studies also indicate that FTNP interferes with the SARS-CoV-2 spike, impairing the cell fusion mechanism. Key points/Highlights • Unique TiO2nanoparticles displaying flavonoid showed potent anti-SARS-CoV-2 activity. • The nanoparticles precisely targeting SARS-CoV-2 were quantitatively verified by cell infectivity in vitro. • Flavonoids on nanoparticles impair the interactions between the spike glycoprotein and ACE-2 receptor. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12112-9.
Collapse
Affiliation(s)
- Gabriela León-Gutiérrez
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico
| | - James Edward Elste
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Carlos Cabello-Gutiérrez
- Departamento de Virología e Investigación en Micología, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, Mexico
| | - Cesar Millán-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Mario H Martínez-Gómez
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico
| | - Rafael Mejía-Alvarez
- Department of Physiology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Armando Mejía
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico.
| |
Collapse
|
3
|
Saultier P, Vidal L, Canault M, Bernot D, Falaise C, Pouymayou C, Bordet JC, Saut N, Rostan A, Baccini V, Peiretti F, Favier M, Lucca P, Deleuze JF, Olaso R, Boland A, Morange PE, Gachet C, Malergue F, Fauré S, Eckly A, Trégouët DA, Poggi M, Alessi MC. Macrothrombocytopenia and dense granule deficiency associated with FLI1 variants: ultrastructural and pathogenic features. Haematologica 2017; 102:1006-1016. [PMID: 28255014 PMCID: PMC5451332 DOI: 10.3324/haematol.2016.153577] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/24/2017] [Indexed: 12/20/2022] Open
Abstract
Congenital macrothrombocytopenia is a family of rare diseases, of which a significant fraction remains to be genetically characterized. To analyze cases of unexplained thrombocytopenia, 27 individuals from a patient cohort of the Bleeding and Thrombosis Exploration Center of the University Hospital of Marseille were recruited for a high-throughput gene sequencing study. This strategy led to the identification of two novel FLI1 variants (c.1010G>A and c.1033A>G) responsible for macrothrombocytopenia. The FLI1 variant carriers’ platelets exhibited a defect in aggregation induced by low-dose adenosine diphosphate (ADP), collagen and thrombin receptor-activating peptide (TRAP), a defect in adenosine triphosphate (ATP) secretion, a reduced mepacrine uptake and release and a reduced CD63 expression upon TRAP stimulation. Precise ultrastructural analysis of platelet content was performed using transmission electron microscopy and focused ion beam scanning electron microscopy. Remarkably, dense granules were nearly absent in the carriers’ platelets, presumably due to a biogenesis defect. Additionally, 25–29% of the platelets displayed giant α-granules, while a smaller proportion displayed vacuoles (7–9%) and autophagosome-like structures (0–3%). In vitro study of megakaryocytes derived from circulating CD34+ cells of the carriers revealed a maturation defect and reduced proplatelet formation potential. The study of the FLI1 variants revealed a significant reduction in protein nuclear accumulation and transcriptional activity properties. Intraplatelet flow cytometry efficiently detected the biomarker MYH10 in FLI1 variant carriers. Overall, this study provides new insights into the phenotype, pathophysiology and diagnosis of FLI1 variant-associated thrombocytopenia.
Collapse
Affiliation(s)
- Paul Saultier
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Léa Vidal
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | | | - Denis Bernot
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Céline Falaise
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Catherine Pouymayou
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | | | - Noémie Saut
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Agathe Rostan
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Véronique Baccini
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | | | - Marie Favier
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Pauline Lucca
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France.,Inserm, UMR_S 1166, Team Genomics and Pathophysiology of Cardiovascular Diseases, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S 1166, France
| | | | - Robert Olaso
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - Anne Boland
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - Pierre Emmanuel Morange
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Christian Gachet
- UMR_S949 INSERM, Strasbourg, France.,Etablissement Français du Sang (EFS)-Alsace, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), France.,Université de Strasbourg, Marseille, France
| | - Fabrice Malergue
- Beckman Coulter Immunotech, Life Sciences Global Assay and Applications Development, Marseille, France
| | - Sixtine Fauré
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Anita Eckly
- UMR_S949 INSERM, Strasbourg, France.,Etablissement Français du Sang (EFS)-Alsace, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), France.,Université de Strasbourg, Marseille, France
| | - David-Alexandre Trégouët
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France.,Inserm, UMR_S 1166, Team Genomics and Pathophysiology of Cardiovascular Diseases, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S 1166, France
| | - Marjorie Poggi
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Marie-Christine Alessi
- Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| |
Collapse
|
4
|
Paris-Trousseau thrombocytopenia is phenocopied by the autosomal recessive inheritance of a DNA-binding domain mutation in FLI1. Blood 2015; 126:2027-30. [PMID: 26316623 DOI: 10.1182/blood-2015-06-650887] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/18/2015] [Indexed: 11/20/2022] Open
Abstract
Hemizygous deletion of a variable region on chromosome 11q containing FLI1 causes an inherited platelet-related bleeding disorder in Paris-Trousseau thrombocytopenia and Jacobsen syndrome. These multisystem disorders are also characterized by heart anomalies, changes in facial structure, and intellectual disability. We have identified a consanguineous family with autosomal recessive inheritance of a bleeding disorder that mimics Paris-Trousseau thrombocytopenia but has no other features of the 11q23 deletion syndrome. Affected individuals in this family have moderate thrombocytopenia; absent collagen-induced platelet aggregation; and large, fused α-granules in 1% to 5% of circulating platelets. This phenotype was caused by a FLI1 homozygous c.970C>T-point mutation that predicts an arginine-to-tryptophan substitution in the conserved ETS DNA-binding domain of FLI1. This mutation caused a transcription defect at the promoter of known FLI1 target genes GP6, GP9, and ITGA2B, as measured by luciferase assay in HEK293 cells, and decreased the expression of these target proteins in affected members of the family as measured by Western blotting of platelet lysates. This kindred suggests abnormalities in FLI1 as causative of Paris-Trousseau thrombocytopenia and confirms the important role of FLI1 in normal platelet development.
Collapse
|
5
|
Kitagawa T, Okita H, Baron B, Tokuda K, Nakamura M, Wang Y, Akada J, Hoshida H, Akada R, Kuramitsu Y, Nakamura K. Mutant screening for oncogenes of Ewing's sarcoma using yeast. Appl Microbiol Biotechnol 2015; 99:6737-44. [PMID: 25936378 DOI: 10.1007/s00253-015-6621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/09/2015] [Accepted: 04/12/2015] [Indexed: 11/30/2022]
Abstract
Many fusion genes, which are the result of chromosomal translocation and work as an oncogene, have been recently identified, but their mode of actions is still unclear. Here, we performed a yeast mutant screening for oncogenes of Ewing's sarcoma to easily identify essential regions responsible for fusion protein functions using a yeast genetic system. Three kinds of oncogenes including EWS/FLI1, EWS/ERG, and EWS/E1AF exhibited growth inhibition in yeast. In this screening, we identified 13 single amino acid substitution mutants which could suppress growth inhibition by oncogenes. All of the point mutation positions of the EWS/ETS family proteins were located within the ETS domain, which is responsible for the interaction with a specific DNA motif. Eight-mutated residues within the ETS domain matched to 13 completely conserved amino acid residues in the human ETS domains. Moreover, mutants also showed reduced transcriptional activities on the DKK2 promoter, which is upregulated by the EWS/ETS family, compared to that of the wild type. These results suggest that the ETS domain in the EWS/ETS family proteins may be a primary target for growth inhibition of Ewing's sarcoma and that this yeast screening system can be applied for the functional screening of the oncogenes.
Collapse
Affiliation(s)
- Takao Kitagawa
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Identification and characterization of nuclear and nucleolar localization signals in the adeno-associated virus serotype 2 assembly-activating protein. J Virol 2014; 89:3038-48. [PMID: 25552709 DOI: 10.1128/jvi.03125-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED Assembly-activating protein (AAP) of adeno-associated virus serotype 2 (AAV2) is a nucleolar-localizing protein that plays a critical role in transporting the viral capsid VP3 protein to the nucleolus for assembly. Here, we identify and characterize AAV2 AAP (AAP2) nuclear (NLS) and nucleolar (NoLS) localization signals near the carboxy-terminal region of AAP2 (amino acid positions 144 to 184) (AAP2(144-184)). This region contains five basic-amino-acid-rich (BR) clusters, KSKRSRR (AAP2BR1), RRR (AAP2BR2), RFR (AAP2BR3), RSTSSR (AAP2BR4), and RRIK (AAP2BR5), from the amino terminus to the carboxy terminus. We created 30 AAP2BR mutants by arginine/lysine-to-alanine mutagenesis or deletion of AAP2BRs and 8 and 1 green fluorescent protein (GFP)-AAP2BR and β-galactosidase-AAP2BR fusion proteins, respectively, and analyzed their intracellular localization in HeLa cells by immunofluorescence microscopy. The results showed that AAP2(144-184) has redundant multipartite NLSs and that any combinations of 4 AAP2BRs, but not 3 or less, can constitute a functional NLS-NoLS; AAP2BR1 and AAP2BR2 play the most influential role for nuclear localization, but either one of the two AAP2BRs is dispensable if all 4 of the other AAP2BRs are present, resulting in 3 different, overlapping NLS motifs; and the NoLS is shared redundantly among the five AAP2BRs and functions in a context-dependent manner. AAP2BR mutations not only resulted in aberrant intracellular localization, but also attenuated AAP2 protein expression to various degrees, and both of these abnormalities have a significant negative impact on capsid production. Thus, this study reveals the organization of the intermingling NLSs and NoLSs in AAP2 and provides insights into their functional roles in capsid assembly. IMPORTANCE Adeno-associated virus (AAV) has become a popular and successful vector for in vivo gene therapy; however, its biology has yet to be fully understood. In this regard, the recent discovery of the assembly-activating protein (AAP), a nonstructural, nucleolar-localizing AAV protein essential for viral capsid assembly, has provided us a new opportunity to better understand the fundamental processes required for virion formation. Here, we identify clusters of basic amino acids in the carboxy terminus of AAP from AAV serotype 2 (AAV2) that act as nuclear and nucleolar localization signals. We also demonstrate their importance in maintaining AAP expression levels and efficient production of viral capsids. Insights into the functions of AAP can elucidate the requirements and process for AAV capsid assembly, which may lead to improved vector production for use in gene therapy. This study also contributes to the growing body of work on nuclear and nucleolar localization signals.
Collapse
|
7
|
Giraud G, Stadhouders R, Conidi A, Dekkers DHW, Huylebroeck D, Demmers JAA, Soler E, Grosveld FG. NLS-tagging: an alternative strategy to tag nuclear proteins. Nucleic Acids Res 2014; 42:gku869. [PMID: 25260593 PMCID: PMC4245968 DOI: 10.1093/nar/gku869] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The characterization of transcription factor complexes and their binding sites in the genome by affinity purification has yielded tremendous new insights into how genes are regulated. The affinity purification requires either the use of antibodies raised against the factor of interest itself or by high-affinity binding of a C- or N-terminally added tag sequence to the factor. Unfortunately, fusing extra amino acids to the termini of a factor can interfere with its biological function or the tag may be inaccessible inside the protein. Here, we describe an effective solution to that problem by integrating the ‘tag’ close to the nuclear localization sequence domain of the factor. We demonstrate the effectiveness of this approach with the transcription factors Fli-1 and Irf2bp2, which cannot be tagged at their extremities without loss of function. This resulted in the identification of novel proteins partners and a new hypothesis on the contribution of Fli-1 to hematopoiesis.
Collapse
Affiliation(s)
- Guillaume Giraud
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ralph Stadhouders
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Andrea Conidi
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Dick H W Dekkers
- Proteomics Center, Erasmus University Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Jeroen A A Demmers
- Proteomics Center, Erasmus University Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Eric Soler
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands Laboratory of Hematopoiesis and Leukemic Stem Cells (LSHL), CEA/INSERM U967, Fontenay-aux-Roses, France Center for Biomedical Genetics and Medical Epigenetics Consortium, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands Center for Biomedical Genetics and Medical Epigenetics Consortium, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands Center for Biomedical Genetics, Erasmus Medical Center, Faculty building, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
8
|
Stockley J, Morgan NV, Bem D, Lowe GC, Lordkipanidzé M, Dawood B, Simpson MA, Macfarlane K, Horner K, Leo VC, Talks K, Motwani J, Wilde JT, Collins PW, Makris M, Watson SP, Daly ME. Enrichment of FLI1 and RUNX1 mutations in families with excessive bleeding and platelet dense granule secretion defects. Blood 2013; 122:4090-3. [PMID: 24100448 PMCID: PMC3862284 DOI: 10.1182/blood-2013-06-506873] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
We analyzed candidate platelet function disorder genes in 13 index cases with a history of excessive bleeding in association with a significant reduction in dense granule secretion and impaired aggregation to a panel of platelet agonists. Five of the index cases also had mild thrombocytopenia. Heterozygous alterations in FLI1 and RUNX1, encoding Friend leukemia integration 1 and RUNT-related transcription factor 1, respectively, which have a fundamental role in megakaryocytopoeisis, were identified in 6 patients, 4 of whom had mild thrombocytopenia. Two FLI1 alterations predicting p.Arg337Trp and p.Tyr343Cys substitutions in the FLI1 DNA-binding domain abolished transcriptional activity of FLI1. A 4-bp deletion in FLI1, and 2 splicing alterations and a nonsense variation in RUNX1, which were predicted to cause haploinsufficiency of either FLI1 or RUNX1, were also identified. Our findings suggest that alterations in FLI1 and RUNX1 may be common in patients with platelet dense granule secretion defects and mild thrombocytopenia.
Collapse
Affiliation(s)
- Jacqueline Stockley
- Department of Cardiovascular Science, University of Sheffield Medical School, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hayashi R, Takeuchi N, Ueda T. Nuclear Respiratory Factor 2β (NRF-2β) recruits NRF-2α to the nucleus by binding to importin-α:β via an unusual monopartite-type nuclear localization signal. J Mol Biol 2013; 425:3536-48. [PMID: 23856623 DOI: 10.1016/j.jmb.2013.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/03/2013] [Accepted: 07/04/2013] [Indexed: 11/28/2022]
Abstract
Nuclear respiratory factor 2 (NRF-2) is a mammalian transcription factor composed of two distinct and unrelated proteins: NRF-2α, which binds to DNA through its Ets domain, and NRF-2β, which contains the transcription activation domain. The activity of NRF-2 in neurons is regulated by nuclear localization; however, the mechanism by which NRF-2 is imported into the nucleus remains unknown. By using in vitro nuclear import assays and immuno-cytofluorescence, we dissect the nuclear import pathways of NRF-2. We show that both NRF-2α and NRF-2β contain intrinsic nuclear localization signals (NLSs): the Ets domain within NRF-2α and the NLS within NRF-2β (amino acids 311/321: EEPPAKRQCIE) that is recognized by importin-α:β. When NRF-2α and NRF-2β form a complex, the nuclear import of NRF-2αβ becomes strictly dependent on the NLS within NRF-2β. Therefore, the nuclear import mechanism of NRF-2 is unique among Ets factors. The NRF-2β NLS contains only two lysine/arginine residues, unlike other known importin-α:β-dependent NLSs. Using ELISA-based binding assays, we show that it is bound by importin-α in almost the same manner and with similar affinity to that of the classical monopartite NLSs, such as c-myc and SV40 T-antigen NLSs. However, the part of the tryptophan array of importin-α that is essential for the recognition of classical monopartite NLSs by generating apolar pockets for the P3 and the P5 lysine/arginine side chains is not required for the recognition of the NRF-2β NLS. We conclude that the NRF-2β NLS is an unusual but is, nevertheless, a bona fide monopartite-type NLS.
Collapse
Affiliation(s)
- Rippei Hayashi
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwanoha, Kashiwa, Chiba Prefecture 277-8562, Japan.
| | | | | |
Collapse
|
10
|
Evans EL, Saxton J, Shelton SJ, Begitt A, Holliday ND, Hipskind RA, Shaw PE. Dimer formation and conformational flexibility ensure cytoplasmic stability and nuclear accumulation of Elk-1. Nucleic Acids Res 2011; 39:6390-402. [PMID: 21543455 PMCID: PMC3159454 DOI: 10.1093/nar/gkr266] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The ETS (E26) protein Elk-1 serves as a paradigm for mitogen-responsive transcription factors. It is multiply phosphorylated by mitogen-activated protein kinases (MAPKs), which it recruits into pre-initiation complexes on target gene promoters. However, events preparatory to Elk-1 phosphorylation are less well understood. Here, we identify two novel, functional elements in Elk-1 that determine its stability and nuclear accumulation. One element corresponds to a dimerization interface in the ETS domain and the second is a cryptic degron adjacent to the serum response factor (SRF)-interaction domain that marks dimerization-defective Elk-1 for rapid degradation by the ubiquitin–proteasome system. Dimerization appears to be crucial for Elk-1 stability only in the cytoplasm, as latent Elk-1 accumulates in the nucleus and interacts dynamically with DNA as a monomer. These findings define a novel role for the ETS domain of Elk-1 and demonstrate that nuclear accumulation of Elk-1 involves conformational flexibility prior to its phosphorylation by MAPKs.
Collapse
Affiliation(s)
- Emma L Evans
- School of Biomedical Sciences, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | |
Collapse
|
11
|
Perdomo J, Fock EL, Kaur G, Yan F, Khachigian LM, Jans DA, Chong BH. A monopartite sequence is essential for p45 NF-E2 nuclear translocation, transcriptional activity and platelet production. J Thromb Haemost 2010; 8:2542-53. [PMID: 20854373 DOI: 10.1111/j.1538-7836.2010.04058.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND p45 NF-E2 is a bZIP transcription factor crucial for thrombopoiesis, as indicated by the fact that loss of p45 NF-E2 function results in dramatic embryonic lethal thrombopoietic defects and its overexpression boosts platelet release. OBJECTIVES In the present study, we set out to identify the sequences responsible for p45 NF-E2 nuclear import, evaluate its transport mechanism and ascertain its functional significance. METHODS A series of p45 NF-E2 deletion constructs fused to green fluorescent protein (GFP) was created and their cellular localization examined in mammalian cells, with the factor responsible for nuclear import identified using an in vitro transport assay. A p45 NF-E2 derivative mutated in the nuclear targeting sequence (NLS) was generated and its biological activity compared with wild type (wt) in luciferase assays, and proplatelet and platelet production measured in murine megakaryocytes transduced with a retroviral vector. RESULTS Here we show that residues 271-273 are essential for nuclear import of p45 NF-E2 in COS-7 and in primary bone marrow cells. The p45 NF-E2 NLS facilitates nuclear import specifically via importin (IMP) 7. Although within the DNA-binding domain of p45 NF-E2, the NLS is not essential for DNA-binding, but is crucial for transcriptional activation and biological activity; where, in contrast to wt, a mutant derivative with a mutated NLS failed to promote proplatelet and platelet production in murine megakaryocytes. CONCLUSIONS The NLS is critical for p45 NF-E2 function, with the present study being the first to demonstrate the importance of NLS-dependent nuclear import of p45 NF-E2 for platelet development.
Collapse
Affiliation(s)
- J Perdomo
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
12
|
Lahaye F, Lespinasse F, Staccini P, Palin L, Paquis-Flucklinger V, Santucci-Darmanin S. hMSH5 is a nucleocytoplasmic shuttling protein whose stability depends on its subcellular localization. Nucleic Acids Res 2010; 38:3655-71. [PMID: 20185565 PMCID: PMC2887964 DOI: 10.1093/nar/gkq098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MSH5 is a MutS-homologous protein required for meiotic DNA recombination. In addition, recent studies suggest that the human MSH5 protein (hMSH5) participates to mitotic recombination and to the cellular response to DNA damage and thus raise the possibility that a tight control of hMSH5 function(s) may be important for genomic stability. With the aim to characterize mechanisms potentially involved in the regulation of hMSH5 activity, we investigated its intracellular trafficking properties. We demonstrate that hMSH5 possesses a CRM1-dependent nuclear export signal (NES) and a nuclear localization signal that participates to its nuclear targeting. Localization analysis of various mutated forms of hMSH5 by confocal microscopy indicates that hMSH5 shuttles between the nucleus and the cytoplasm. We also provide evidence suggesting that hMSH5 stability depends on its subcellular compartmentalization, hMSH5 being much less stable in the nucleus than in the cytoplasm. Together, these data suggest that hMSH5 activity may be regulated by nucleocytoplasmic shuttling and nuclear proteasomal degradation, both of these mechanisms contributing to the control of nuclear hMSH5 content. Moreover, data herein also support that in tissues where both hMSH5 and hMSH4 proteins are expressed, hMSH5 might be retained in the nucleus through masking of its NES by binding of hMSH4.
Collapse
Affiliation(s)
- François Lahaye
- FRE 3086 Instabilité génétique: Maladies rares et cancers, Université de Nice Sophia-Antipolis, CNRS, Nice Cedex 2, France
| | | | | | | | | | | |
Collapse
|
13
|
Okamura K, Yamashita S, Ando H, Horibata Y, Aoyama C, Takagishi K, Izumi T, Vance DE, Sugimoto H. Identification of nuclear localization and nuclear export signals in Ets2, and the transcriptional regulation of Ets2 and CTP:phosphocholine cytidylyltransferase α in tetradecanoyl-13-acetate or macrophage-colony stimulating factor stimulated RAW264 cells. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:173-82. [DOI: 10.1016/j.bbalip.2008.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 12/16/2008] [Accepted: 12/18/2008] [Indexed: 11/24/2022]
|
14
|
Meade AJ, Meloni BP, Mastaglia FL, Knuckey NW. The application of cell penetrating peptides for the delivery of neuroprotective peptides/proteins in experimental cerebral ischaemia studies. ACTA ACUST UNITED AC 2009. [DOI: 10.6030/1939-067x-2.1.21] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
15
|
Kwok JC, Perdomo J, Chong BH. Identification of a monopartite sequence in PU.1 essential for nuclear import, DNA-binding and transcription of myeloid-specific genes. J Cell Biochem 2008; 101:1456-74. [PMID: 17340619 DOI: 10.1002/jcb.21264] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Ets transcription factor PU.1 is an essential regulator of normal hematopoiesis, especially within the myeloid lineage. As such, endogenous PU.1 predominantly localizes to the nucleus of mammalian cells to facilitate gene regulation. However, to date, little is known regarding the mechanisms of PU.1 nuclear transport. We found, using HeLa and RAW 264.7 macrophage cells, that PU.1 enters the nucleus via passive diffusion and active transport. The latter can be facilitated by: (i) the classical pathway requiring importin alpha and beta; (ii) the non-classical pathway requiring only importin beta; or (iii) direct interaction with nucleoporins. A group of six positively charged lysine or arginine residues within the Ets DNA-binding domain was determined to be crucial in active nuclear import. These residues directly interact with importin beta to facilitate a predominantly non-classical import pathway. Furthermore, luciferase reporter assays demonstrated that these same six amino acids are crucial for PU.1-mediated transcriptional activation of myeloid-specific genes. Indeed, these residues may represent a consensus sequence vital for nuclear import, DNA-binding and transcriptional activity of Ets family members. By identifying and characterizing the mechanisms of PU.1 nuclear import and the specific amino acids involved, this report may provide insights into the molecular basis of diseases.
Collapse
Affiliation(s)
- Juliana C Kwok
- Centre for Thrombosis and Vascular Research, Department of Medicine, St. George Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | | | | |
Collapse
|
16
|
Theodore M, Kawai Y, Yang J, Kleshchenko Y, Reddy SP, Villalta F, Arinze IJ. Multiple nuclear localization signals function in the nuclear import of the transcription factor Nrf2. J Biol Chem 2008; 283:8984-94. [PMID: 18238777 DOI: 10.1074/jbc.m709040200] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) mediates the transcriptional response of cells to oxidative stress and is translocated into the nucleus following, or concomitant with, its activation by electrophiles or reactive oxygen species. The mechanism of its translocation into the nucleus is not entirely elucidated. Here we have identified two novel nuclear localization signal (NLS) motifs in murine Nrf2, one located near the N-terminal region (amino acid residues 42-53) and the other (residues 587-593) located near the C-terminal region. Imaging of green fluorescent protein (GFP)-tagged Nrf2 revealed that mutation(s) in any of these sequences resulted in decreased nuclear fluorescence intensity compared with the wild-type Nrf2 when Nrf2 activation was induced with the electrophile tert-butylhydroquinone. The mutations also impaired Nrf2-induced transactivation of antioxidant response element-driven reporter gene expression to the same extent as the Nrf2 construct bearing mutation in a previously identified bipartite NLS that maps at residues 494-511. When linked to GFP or to GFP-PEPCK-C each of the novel NLS motifs was sufficient to drive nuclear translocation of the fusion proteins. Co-immunoprecipitation assays demonstrated that importins alpha5 and beta1 associate with Nrf2, an interaction that was blocked by the nuclear import inhibitor SN50. SN50 also blocked tert-butylhydroquinone-induced nuclear fluorescence of GFP-Nrf2 in cells transfected with wild-type GFP-Nrf2. Overall these results reveal that multiple NLS motifs in Nrf2 function in its nuclear translocation in response to pro-oxidant stimuli and that the importin alpha-beta heterodimer nuclear import receptor system plays a critical role in the import process.
Collapse
Affiliation(s)
- Melanie Theodore
- School of Medicine, Meharry Medical College, Nashville, TN 37208-3599, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Philips AS, Kwok JC, Chong BH. Analysis of the signals and mechanisms mediating nuclear trafficking of GATA-4. Loss of DNA binding is associated with localization in intranuclear speckles. J Biol Chem 2007; 282:24915-27. [PMID: 17548362 DOI: 10.1074/jbc.m701789200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nucleocytoplasmic transport of GATA-4 is important in maintaining and regulating normal cardiogenesis and heart function. This report investigates the detailed mechanisms of GATA-4 nuclear transport. We characterized a nonclassical nuclear localization signal between amino acids 270 and 324 that actively transports GATA-4 into the nucleus of both HeLa cells and cardiac myocytes. Fine mapping studies revealed four crucial arginine residues within this region that mediate active transport predominantly through the nonclassical pathway via interaction with importin beta. These four residues were also essential for the DNA binding activity of GATA-4 and transcriptional activation of cardiac-specific genes. Interestingly, mutation of these residues not only inhibited DNA binding and gene transcription but also resulted in a preferential accumulation of the GATA-4 protein in distinct subnuclear speckles. A cardiac myocyte-specific, chromosome maintenance region 1-dependent nuclear export signal consisting of three essential leucine residues was also identified. The current study provides detailed information on the nuclear shuttling pathways of GATA-4 that represents an additional mechanism of gene regulation.
Collapse
Affiliation(s)
- Alana S Philips
- Centre for Vascular Research, Department of Medicine, St. George Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | | | |
Collapse
|
18
|
Abstract
The ETS (E26 Transformation-specific Sequence) factors are comprised of a family of transcription factors that share a highly conserved DNA binding domain. Although originally described for their role as protooncogenes in the development of several types of human cancer, they have subsequently been shown to regulate a wide variety of biological processes including cellular growth and differentiation under normal and pathological conditions. As transcription factors, they can either function as activators or repressors of gene expression. Several ETS family members are expressed in cells of vascular origin, including endothelial cells and vascular smooth muscle cells, where they regulate the expression of a number of vascular-specific genes. In the past few years, emerging evidence supports a novel role for selected ETS family members in the regulation of vascular inflammation and remodeling. ETS factor expression can be induced by proinflammatory cytokines, growth factors, and vasoactive peptides. Examples of some of the target genes regulated by ETS factors include adhesion molecules, chemokines, and matrix metalloproteinases. Targeted disruption of selected ETS family members such as Ets-1 in mice is associated with marked reductions in the recruitment of inflammatory cells and vascular remodeling in response to systemic administration of the vasoactive peptide angiotensin II. The purpose of this review is to provide an overview of recent advances that have been made in defining a role for selected members of the ETS transcription factor family in the regulation of vascular-specific gene expression, vascular inflammation, and remodeling.
Collapse
Affiliation(s)
- Peter Oettgen
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA.
| |
Collapse
|
19
|
|
20
|
Do HJ, Song H, Yang HM, Kim DK, Kim NH, Kim JH, Cha KY, Chung HM, Kim JH. Identification of multiple nuclear localization signals in murine Elf3, an ETS transcription factor. FEBS Lett 2006; 580:1865-71. [PMID: 16516205 DOI: 10.1016/j.febslet.2006.02.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 02/17/2006] [Accepted: 02/17/2006] [Indexed: 11/21/2022]
Abstract
We investigated nuclear localization signal (NLS) determinants within the AT-hook and ETS DNA-binding domains of murine Elf3 (mElf3), a member of the subfamily of epithelium-specific ETS transcription factors. Deletion mutants containing the AT-hook, ETS domain or both localized strictly in the nucleus, suggesting that these individual domains contain independent NLS motif(s). Within the AT-hook domain, four basic residues (244KRKR247) were critical for strong NLS activity, and two potent bipartite NLS motifs (236-252 and 249-267) were sufficient for nuclear import of mElf3, although less efficient than the full domain. In addition, one stretch of basic residues (318KKK320) within the ETS domain appears to be essential for mElf3 nuclear localization. Taken together, mElf3 contains multiple NLS motifs, which may function cooperatively to effect efficient nuclear transport.
Collapse
Affiliation(s)
- Hyun-Jin Do
- Chabiotech Co. Ltd., Seoul 135-907, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Camuzeaux B, Spriet C, Héliot L, Coll J, Duterque-Coquillaud M. Imaging Erg and Jun transcription factor interaction in living cells using fluorescence resonance energy transfer analyses. Biochem Biophys Res Commun 2005; 332:1107-14. [PMID: 15922298 DOI: 10.1016/j.bbrc.2005.05.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Accepted: 05/05/2005] [Indexed: 10/25/2022]
Abstract
Physical interactions between transcription factors play important roles in modulating gene expression. Previous in vitro studies have shown a transcriptional synergy between Erg protein, an Ets family member, and Jun/Fos heterodimer, members of the bZip family, which requires direct Erg-Jun protein interactions. Visualization of protein interactions in living cells is a new challenge in biology. For this purpose, we generated fusion proteins of Erg, Fos, and Jun with yellow and cyan fluorescent proteins, YFP and CFP, respectively. After transient expression in HeLa cells, interactions of the resulting fusion proteins were explored by fluorescence resonance energy transfer microscopy (FRET) in fixed and living cells. FRET between YFP-Erg and CFP-Jun was monitored by using photobleaching FRET and fluorescence lifetime imaging microscopy. Both techniques revealed the occurrence of intermolecular FRET between YFP-Erg and CFP-Jun. This is stressed by loss of FRET with an YFP-Erg version carrying a point mutation in its ETS domain. These results provide evidence for the interaction of Erg and Jun proteins in living cells as a critical prerequisite of their transcriptional synergy, but also for the essential role of the Y371 residue, conserved in most Ets proteins, in this interaction.
Collapse
Affiliation(s)
- Barbara Camuzeaux
- UMR 8526CNRS/Institut Pasteur de Lille/Université de Lille2, Institut de Biologie de Lille, BP 447, 1 rue Calmette, 59021 Lille cedex, France
| | | | | | | | | |
Collapse
|