1
|
Zhang H, Xue F, Zhao H, Chen L, Wang T, Wu X. DNA methylation status of DNAJA4 is essential for human erythropoiesis. Epigenomics 2022; 14:1249-1267. [DOI: 10.2217/epi-2022-0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Aims: To investigate DNA methylation patterns in early and terminal stages of erythropoiesis, and to explore the function of differentially methylated genes in erythropoiesis and erythroid disorders. Materials & methods: Differential analysis of DNA methylation and gene expression during erythropoiesis, as well as weighted gene coexpression network analysis of acute myeloid leukemia was performed. Results: We identified four candidate genes that possessed differential methylation in the promoter regions. DNAJA4 affected proliferation, apoptosis and enucleation during terminal erythropoiesis and was associated with the prognosis of acute myeloid leukemia. DNAJA4 was specifically highly expressed in erythroleukemia and is associated with DNA methylation. Conclusion: DNAJA4 plays a crucial role for erythropoiesis and is regulated via DNA methylation. Dysregulation of DNAJA4 expression is associated with erythroid disorders.
Collapse
Affiliation(s)
- Hengchao Zhang
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Fumin Xue
- Department of Gastroenterology, Children’s Hospital affiliated of Zhengzhou University, Zhengzhou, 450000, China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Ting Wang
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Xiuyun Wu
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| |
Collapse
|
2
|
Mohamed RA, Guo CT, Xu SY, Ying SH, Feng MG. Characterization of BbKlf1 as a novel transcription factor vital for asexual and infection cycles of Beauveria bassiana. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:719-731. [PMID: 35851566 DOI: 10.1111/1758-2229.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
The large family of C2H2-type zinc finger transcription factors (TFs) comprise the Kruppel-like factors (KLFs) that evolved relatively late in eukaryotes but remain unexplored in filamentous fungi. Here, we report that an orthologue (BbKlf1) of yeast Klf1 mediating cell wall integrity (CWI) is a wide-spectrum TF evidently localized in nucleus and cytoplasm in Beauveria bassiana. BbKlf1 features conserved domains and multiple DNA-binding motifs predicted to bind multiple promoter DNA fragments of target genes across asexual developmental and stress-responsive pathways. Despite limited impact on normal colony growth, deletion of Bbklf1 resulted in impaired CWI and hypersensitivity to Congo red-induced cell wall stress. Also, the deletion mutant was severely compromised in tolerance to oxidative and osmotic stresses, hyphal septation and differentiation, conidiation capacity (reduced by 95%), conidial quality (viability and hydrocarbon epitope pattern) and virulence. Importantly, these phenotypes correlated well with sharply repressed or nearly abolished expressions of those genes required for or involved in chitin biosynthesis, antioxidant activity, cell division and differentiation, aerial conidiation and conidial maturation. These findings indicate an essentiality of BbKlf1 for the asexual and insect-pathogenic lifecycles of B. bassiana and a novel scenario much beyond the yeast orthologue-mediated CWI, suggesting important roles of its orthologues in filamentous fungi.
Collapse
Affiliation(s)
- Rehab Abdelmonem Mohamed
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chong-Tao Guo
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Si-Yuan Xu
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng-Hua Ying
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ming-Guang Feng
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Mansoor A, Mansoor MO, Patel JL, Zhao S, Natkunam Y, Bieker JJ. KLF1/EKLF expression in acute leukemia is correlated with chromosomal abnormalities. Blood Cells Mol Dis 2020; 83:102434. [PMID: 32311573 DOI: 10.1016/j.bcmd.2020.102434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022]
Abstract
KLF1 (EKLF) is a master regulator of erythropoiesis and controls expression of a wide array of target genes. We interrogated human tissue microarray samples via immunohistological analysis to address whether levels of KLF1 protein are associated with leukemia. We have made the unexpected findings that higher KLF1 levels are correlated with cells containing abnormal chromosomes, and that high KLF1 expression is not limited to acute myeloid leukemia (AML) associated with erythroid/megakaryoblastic differentiation. Expression of KLF1 is associated with poor survival. Further analyses reveal that KLF1 directly regulates a number of genes that play a role in chromosomal integrity. Together these results suggest that monitoring KLF1 levels may provide a new marker for risk stratification and prognosis in patients with AML.
Collapse
Affiliation(s)
- Adnan Mansoor
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Mohammad Omer Mansoor
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Jay L Patel
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - James J Bieker
- Department of Cell, Developmental, & Regenerative Biology, Black Family Stem Cell Institute, Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Gnanapragasam MN, Crispino JD, Ali AM, Weinberg R, Hoffman R, Raza A, Bieker JJ. Survey and evaluation of mutations in the human KLF1 transcription unit. Sci Rep 2018; 8:6587. [PMID: 29700354 PMCID: PMC5920080 DOI: 10.1038/s41598-018-24962-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023] Open
Abstract
Erythroid Krüppel-like Factor (EKLF/KLF1) is an erythroid-enriched transcription factor that plays a global role in all aspects of erythropoiesis, including cell cycle control and differentiation. We queried whether its mutation might play a role in red cell malignancies by genomic sequencing of the KLF1 transcription unit in cell lines, erythroid neoplasms, dysplastic disorders, and leukemia. In addition, we queried published databases from a number of varied sources. In all cases we only found changes in commonly notated SNPs. Our results suggest that if there are mutations in KLF1 associated with erythroid malignancies, they are exceedingly rare.
Collapse
Affiliation(s)
- Merlin Nithya Gnanapragasam
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - John D Crispino
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Abdullah M Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Rona Weinberg
- Cellular Therapy Laboratory, New York Blood Center, New York, NY, 10065, USA
| | - Ronald Hoffman
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - Azra Raza
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Black Familly Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
- Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA.
| |
Collapse
|
5
|
Gillinder KR, Ilsley MD, Nébor D, Sachidanandam R, Lajoie M, Magor GW, Tallack MR, Bailey T, Landsberg MJ, Mackay JP, Parker MW, Miles LA, Graber JH, Peters LL, Bieker JJ, Perkins AC. Promiscuous DNA-binding of a mutant zinc finger protein corrupts the transcriptome and diminishes cell viability. Nucleic Acids Res 2017; 45:1130-1143. [PMID: 28180284 PMCID: PMC5388391 DOI: 10.1093/nar/gkw1014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/13/2016] [Accepted: 11/02/2016] [Indexed: 12/27/2022] Open
Abstract
The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.
Collapse
Affiliation(s)
- Kevin R Gillinder
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Melissa D Ilsley
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | | | - Ravi Sachidanandam
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Mathieu Lajoie
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Graham W Magor
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Michael R Tallack
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Timothy Bailey
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, USA
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, NSW, Australia
| | - Michael W Parker
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia.,ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Luke A Miles
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, USA
| | - Andrew C Perkins
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia.,Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| |
Collapse
|
6
|
Ilsley MD, Gillinder KR, Magor GW, Huang S, Bailey TL, Crossley M, Perkins AC. Krüppel-like factors compete for promoters and enhancers to fine-tune transcription. Nucleic Acids Res 2017; 45:6572-6588. [PMID: 28541545 PMCID: PMC5499887 DOI: 10.1093/nar/gkx441] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Krüppel-like factors (KLFs) are a family of 17 transcription factors characterized by a conserved DNA-binding domain of three zinc fingers and a variable N-terminal domain responsible for recruiting cofactors. KLFs have diverse functions in stem cell biology, embryo patterning, and tissue homoeostasis. KLF1 and related family members function as transcriptional activators via recruitment of co-activators such as EP300, whereas KLF3 and related members act as transcriptional repressors via recruitment of C-terminal Binding Proteins. KLF1 directly activates the Klf3 gene via an erythroid-specific promoter. Herein, we show KLF1 and KLF3 bind common as well as unique sites within the erythroid cell genome by ChIP-seq. We show KLF3 can displace KLF1 from key erythroid gene promoters and enhancers in vivo. Using 4sU RNA labelling and RNA-seq, we show this competition results in reciprocal transcriptional outputs for >50 important genes. Furthermore, Klf3-/- mice displayed exaggerated recovery from anemic stress and persistent cell cycling consistent with a role for KLF3 in dampening KLF1-driven proliferation. We suggest this study provides a paradigm for how KLFs work in incoherent feed-forward loops or networks to fine-tune transcription and thereby control diverse biological processes such as cell proliferation.
Collapse
Affiliation(s)
- Melissa D. Ilsley
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
| | - Kevin R. Gillinder
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
| | - Graham W. Magor
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
| | - Stephen Huang
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
| | | | | | - Andrew C. Perkins
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
- The Princess Alexandra Hospital, Brisbane 4102, Australia
| |
Collapse
|
7
|
Ghaleb AM, Yang VW. Krüppel-like factor 4 (KLF4): What we currently know. Gene 2017; 611:27-37. [PMID: 28237823 DOI: 10.1016/j.gene.2017.02.025] [Citation(s) in RCA: 390] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 02/06/2023]
Abstract
Krüppel-like factor 4 (KLF4) is an evolutionarily conserved zinc finger-containing transcription factor that regulates diverse cellular processes such as cell growth, proliferation, and differentiation. Since its discovery in 1996, KLF4 has been gaining a lot of attention, particularly after it was shown in 2006 as one of four factors involved in the induction of pluripotent stem cells (iPSCs). Here we review the current knowledge about the different functions and roles of KLF4 in various tissue and organ systems.
Collapse
Affiliation(s)
- Amr M Ghaleb
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Vincent W Yang
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW The nature and function of macrophages at the center of erythroblastic islands is not fully understood. This review discusses novel findings on the phenotypic and molecular characterization of erythroblastic island macrophages, and their role in regulating normal and pathological erythropoiesis. RECENT FINDINGS The phenotype to prospectively isolate erythroblastic island macrophages from mouse bone marrow has been identified. In-vivo depletion of erythroblastic island macrophages causes blockade of erythroblast maturation and delays erythropoietic recovery following chemical insults. The cytokine granulocyte colony-stimulating factor arrests medullary erythropoiesis by depleting erythroblastic island macrophages from the bone marrow. In-vivo ablation of macrophages improves anemia associated with β-thalassemia and reduces red blood cell counts in the mouse model of polycythemia vera. The role of cell adhesion molecules regulating interactions between erythroblastic island macrophages and erythroblasts has been clarified, and mechanisms of pyrenocyte engulfment by erythroblastic island macrophages have been demonstrated to involve Mer tyrosine kinase receptor. SUMMARY Prospective isolation of mouse erythroblastic island macrophages together with new genetic mouse models to specifically target erythroblastic island macrophages will enable molecular studies to better define their role in controlling erythroblast maturation. These studies have revealed the key role of erythroblastic island macrophages in regulating normal erythropoiesis and could be interesting targets to treat β-thalassemia or polycythemia vera.
Collapse
|
9
|
Dobrivojević M, Habek N, Kapuralin K, Ćurlin M, Gajović S. Krüppel-like transcription factor 8 (Klf8) is expressed and active in the neurons of the mouse brain. Gene 2015; 570:132-40. [PMID: 26071188 DOI: 10.1016/j.gene.2015.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/27/2015] [Accepted: 06/04/2015] [Indexed: 10/23/2022]
Abstract
Krüppel-like transcription factor 8 (KLF8) is a transcription factor suggested to be involved in various cellular events, including malignant cell transformation, still its expression in the adult rodent brain remained unknown. To analyze Klf8 in the mouse brain and to identify cell types expressing it, a specific transgenic Klf8(Gt1Gaj) mouse was used. The resulting Klf8 gene-driven β-galactosidase activity was visualized by X-gal histochemical staining of the brain sections. The obtained results were complemented by in situ RNA hybridization and immunohistochemistry. Klf8 was highly expressed throughout the adult mouse brain gray matter including the cerebral cortex, hippocampus, olfactory bulb, hypothalamus, pallidum, and striatum, but not in the cerebellum. Immunofluorescent double-labeling revealed that KLF8-immunoreactive cells were neurons, and the staining was located in their nucleus. This was the first study showing that Klf8 was highly expressed in various regions of the mouse brain and in particular in the neurons, where it was localized in the cell nuclei.
Collapse
Affiliation(s)
- Marina Dobrivojević
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia
| | - Nikola Habek
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia
| | - Katarina Kapuralin
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia
| | - Marija Ćurlin
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia
| | - Srećko Gajović
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia.
| |
Collapse
|
10
|
Maksimenko O, Georgiev P. Mechanisms and proteins involved in long-distance interactions. Front Genet 2014; 5:28. [PMID: 24600469 PMCID: PMC3927085 DOI: 10.3389/fgene.2014.00028] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 01/25/2014] [Indexed: 12/28/2022] Open
Abstract
Due to advances in genome-wide technologies, consistent distant interactions within chromosomes of higher eukaryotes have been revealed. In particular, it has been shown that enhancers can specifically and directly interact with promoters by looping out intervening sequences, which can be up to several hundred kilobases long. This review is focused on transcription factors that are supposed to be involved in long-range interactions. Available data are in agreement with the model that several known transcription factors and insulator proteins belong to an abundant but poorly studied class of proteins that are responsible for chromosomal architecture.
Collapse
Affiliation(s)
- Oksana Maksimenko
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences Moscow, Russia
| | - Pavel Georgiev
- Department of the Control of Genetic Processes, Institute of Gene Biology, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
11
|
Yien YY, Bieker JJ. EKLF/KLF1, a tissue-restricted integrator of transcriptional control, chromatin remodeling, and lineage determination. Mol Cell Biol 2013; 33:4-13. [PMID: 23090966 PMCID: PMC3536305 DOI: 10.1128/mcb.01058-12] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Erythroid Krüppel-like factor (EKLF or KLF1) is a transcriptional regulator that plays a critical role in lineage-restricted control of gene expression. KLF1 expression and activity are tightly controlled in a temporal and differentiation stage-specific manner. The mechanisms by which KLF1 is regulated encompass a range of biological processes, including control of KLF1 RNA transcription, protein stability, localization, and posttranslational modifications. Intact KLF1 regulation is essential to correctly regulate erythroid function by gene transcription and to maintain hematopoietic lineage homeostasis by ensuring a proper balance of erythroid/megakaryocytic differentiation. In turn, KLF1 regulates erythroid biology by a wide variety of mechanisms, including gene activation and repression by regulation of chromatin configuration, transcriptional initiation and elongation, and localization of gene loci to transcription factories in the nucleus. An extensive series of biochemical, molecular, and genetic analyses has uncovered some of the secrets of its success, and recent studies are highlighted here. These reveal a multilayered set of control mechanisms that enable efficient and specific integration of transcriptional and epigenetic controls and that pave the way for proper lineage commitment and differentiation.
Collapse
Affiliation(s)
- Yvette Y. Yien
- Department of Developmental and Regenerative Biology
- Graduate School of Biological Sciences
| | - James J. Bieker
- Department of Developmental and Regenerative Biology
- Black Family Stem Cell Institute
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
12
|
Brancaleoni V, Granata F, Colancecco A, Tavazzi D, Cappellini MD, Di Pierro E. Seven novel genetic mutations within the 5'UTR and the housekeeping promoter of HMBS gene responsible for the non-erythroid form of acute intermittent porphyria. Blood Cells Mol Dis 2012; 49:147-51. [PMID: 22748422 DOI: 10.1016/j.bcmd.2012.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 06/03/2012] [Indexed: 11/17/2022]
Abstract
Acute intermittent porphyria (AIP) is an autosomal dominant disorder caused by molecular abnormalities in the HMBS gene. This gene is transcribed from two promoters to produce ubiquitous and erythroid specific isoforms of porphobilinogen deaminase (PBGD). In the classical form of AIP, both isoforms are deficient, but about 5% of families have the non-erythroid variant in which only the ubiquitous isoform is affected. Only one mutation sited in the housekeeping promoter has been previously reported as causative for this form of AIP. In this study, we identified one small deletion and six nucleotide substitutions within the 5'UTR and the housekeeping promoter of HMBS gene: c.1-440_-427del14bp; c.1-421G>A; c.1-331C>T; c.1-270G>A; c.1-122T>A; c.1-103C>T; c.1-28A>C. Using luciferase reporter assays and quantitative PCR experiments, we characterized the functional role of these seven novel genetic variants demonstrating that all mutations cause a significant loss of transcriptional activity. Our investigations suggest that these nucleotide substitutions may alter critical binding sites for transcriptional factors, which confirms that these regions represent an important molecular target for pathogenesis of non-erythroid form of acute intermittent porphyria.
Collapse
Affiliation(s)
- Valentina Brancaleoni
- Dipartimento di Medicina Interna, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
The cellular events that lead to terminal erythroid differentiation rely on the controlled interplay of extra- and intracellular regulatory factors. Their downstream effects are highly coordinated and result in the structural/morphologic and metabolic changes that uniquely characterize a maturing red blood cell. Erythroid Krüppel-like factor (EKLF/KLF1) is one of a very small number of intrinsic transcription factors that play a major role in regulating these events. This review covers 3 major aspects of erythropoiesis in which EKLF plays crucial functions: (1) at the megakaryocyte-erythroid progenitor stage, where it is involved in erythroid lineage commitment; (2) during the global expansion of erythroid gene expression in primitive and definitive lineages, where it plays a direct role in globin switching; and (3) during the terminal maturation of red cells, where it helps control exit from the cell cycle. We conclude by describing recent studies of mammalian EKLF/KLF1 mutations that lead to altered red cell phenotypes and disease.
Collapse
Affiliation(s)
- Miroslawa Siatecka
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
14
|
Tallack MR, Perkins AC. KLF1 directly coordinates almost all aspects of terminal erythroid differentiation. IUBMB Life 2011; 62:886-90. [PMID: 21190291 DOI: 10.1002/iub.404] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The molecular events and transcriptional mechanisms that underlie erythropoiesis are of great interest to biologists and hematologists since disorders of erythrocytes are common and remain relatively poorly understood. Kruppel-like factor 1 (KLF1) is a critical transcription factor for erythropoiesis in mice and man. Recently the use of chromatin immunoprecipitation (ChIP) coupled to next-generation DNA sequencing (ChIP-seq) has led to an updated understanding of how KLF1 functions in vivo. The full extent of KLF1 target genes have provided new insights into erythropoiesis, and have established that KLF1 controls almost all aspects of erythroid cell development and maturation.
Collapse
Affiliation(s)
- Michael R Tallack
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
15
|
Abstract
The transition from fetal to adult beta-like globin expression is a key step in the maturation of the red blood cell lineage. Two new studies show that the KLF1 zinc finger protein uses direct and indirect means to regulate the final switch from fetal to adult globin expression and that monoallelic loss of KLF1 expression leads to persistence of fetal hemoglobin.
Collapse
Affiliation(s)
- James J Bieker
- Mount Sinai School of Medicine, Department of Developmental and Regenerative Biology, New York, New York, USA
| |
Collapse
|
16
|
Severe anemia in the Nan mutant mouse caused by sequence-selective disruption of erythroid Kruppel-like factor. Proc Natl Acad Sci U S A 2010; 107:15151-6. [PMID: 20696915 DOI: 10.1073/pnas.1004996107] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies of mouse models of anemia have long provided fundamental insights into red blood cell formation and function. Here we show that the semidominant mouse mutation Nan ("neonatal anemia") carries a single amino acid change (E339D) within the second zinc finger of the erythroid Krüppel-like factor (EKLF), a critical erythroid regulatory transcription factor. The mutation alters the DNA-binding specificity of EKLF so that it no longer binds promoters of a subset of its DNA targets. Remarkably, even when mutant Nan and wild-type EKLF alleles are expressed at equivalent levels, the mutant form selectively interferes with expression of EKLF target genes whose promoter elements it no longer binds. This interference yields a distorted genetic output and selective protein deficiencies that differ from those seen in EKLF-heterozygous and EKLF-null red blood cells and presents a unique and unexpected mechanism of inherited disease.
Collapse
|
17
|
Tallack MR, Whitington T, Yuen WS, Wainwright EN, Keys JR, Gardiner BB, Nourbakhsh E, Cloonan N, Grimmond SM, Bailey TL, Perkins AC. A global role for KLF1 in erythropoiesis revealed by ChIP-seq in primary erythroid cells. Genome Res 2010; 20:1052-63. [PMID: 20508144 DOI: 10.1101/gr.106575.110] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
KLF1 regulates a diverse suite of genes to direct erythroid cell differentiation from bipotent progenitors. To determine the local cis-regulatory contexts and transcription factor networks in which KLF1 operates, we performed KLF1 ChIP-seq in the mouse. We found at least 945 sites in the genome of E14.5 fetal liver erythroid cells which are occupied by endogenous KLF1. Many of these recovered sites reside in erythroid gene promoters such as Hbb-b1, but the majority are distant to any known gene. Our data suggests KLF1 directly regulates most aspects of terminal erythroid differentiation including production of alpha- and beta-globin protein chains, heme biosynthesis, coordination of proliferation and anti-apoptotic pathways, and construction of the red cell membrane and cytoskeleton by functioning primarily as a transcriptional activator. Additionally, we suggest new mechanisms for KLF1 cooperation with other transcription factors, in particular the erythroid transcription factor GATA1, to maintain homeostasis in the erythroid compartment.
Collapse
Affiliation(s)
- Michael R Tallack
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Tsiftsoglou AS, Vizirianakis IS, Strouboulis J. Erythropoiesis: model systems, molecular regulators, and developmental programs. IUBMB Life 2009; 61:800-30. [PMID: 19621348 DOI: 10.1002/iub.226] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human erythropoiesis is a complex multistep developmental process that begins at the level of pluripotent hematopoietic stem cells (HSCs) at bone marrow microenvironment (HSCs niche) and terminates with the production of erythrocytes (RBCs). This review covers the basic and contemporary aspects of erythropoiesis. These include the: (a) cell-lineage restricted pathways of differentiation originated from HSCs and going downward toward the blood cell development; (b) model systems employed to study erythropoiesis in culture (erythroleukemia cell lines and embryonic stem cells) and in vivo (knockout animals: avian, mice, zebrafish, and xenopus); (c) key regulators of erythropoiesis (iron, hypoxia, stress, and growth factors); (d) signaling pathways operating at hematopoietic stem cell niche for homeostatic regulation of self renewal (SCF/c-kit receptor, Wnt, Notch, and Hox) and for erythroid differentiation (HIF and EpoR). Furthermore, this review presents the mechanisms through which transcriptional factors (GATA-1, FOG-1, TAL-1/SCL/MO2/Ldb1/E2A, EKLF, Gfi-1b, and BCL11A) and miRNAs regulate gene pattern expression during erythroid differentiation. New insights regarding the transcriptional regulation of alpha- and beta-globin gene clusters were also presented. Emphasis was also given on (i) the developmental program of erythropoiesis, which consists of commitment to terminal erythroid maturation and hemoglobin production, (two closely coordinated events of erythropoieis) and (ii) the capacity of human embryonic and umbilical cord blood (UCB) stem cells to differentiate and produce RBCs in culture with highly selective media. These most recent developments will eventually permit customized red blood cell production needed for transfusion.
Collapse
Affiliation(s)
- Asterios S Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | |
Collapse
|
19
|
Ryu MS, Lichten LA, Liuzzi JP, Cousins RJ. Zinc transporters ZnT1 (Slc30a1), Zip8 (Slc39a8), and Zip10 (Slc39a10) in mouse red blood cells are differentially regulated during erythroid development and by dietary zinc deficiency. J Nutr 2008; 138:2076-83. [PMID: 18936201 DOI: 10.3945/jn.108.093575] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Zinc is essential for normal erythroid cell functions and therefore intracellular zinc homeostasis during erythroid differentiation is tightly regulated. However, a characterization of zinc transporters in erythrocytes has not been conducted. The membrane fraction of mature mouse RBC was screened for zinc transporter expression using western analysis as a first step in the characterization process. ZnT1, Zip8, and Zip10 were detected among the 12 transporter proteins tested. We examined expression of these zinc transporters during erythropoietin (EPO)-induced differentiation of splenic erythroid progenitor cells into reticulocytes. Both Zip8 and Zip10 mRNA increased by 2-6 h after addition of EPO to the cells. In contrast, maximal RNA levels for the zinc transporter ZnT1 and erythroid delta-aminolevulinic acid synthase were only produced by 24 h after EPO. We confirmed these changes in transcript abundance by western analysis. Dietary zinc status influences zinc-dependent functions of RBC. To determine whether the identified zinc transporters respond to dietary zinc status, mice were fed a zinc-deficient or control diet. Incorporation of (65)Zn into erythrocytes in vitro was significantly increased in cells from the zinc-deficient mice. Western analysis and densitometry revealed that erythrocyte Zip10 was upregulated and ZnT1 was downregulated in the zinc-depleted mice. Zip8 was not affected by restricted zinc intake. Collectively, these data suggest that the zinc transporters ZnT1, Zip8, and Zip10 are important for zinc homeostasis in erythrocytes and that ZnT1 and Zip10 respond to the dietary zinc supply.
Collapse
Affiliation(s)
- Moon-Suhn Ryu
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-0370, USA
| | | | | | | |
Collapse
|
20
|
EKLF restricts megakaryocytic differentiation at the benefit of erythrocytic differentiation. Blood 2008; 112:576-84. [DOI: 10.1182/blood-2007-07-098996] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Abstract
Previous observations suggested that functional antagonism between FLI-1 and EKLF might be involved in the commitment toward erythrocytic or megakaryocytic differentiation. We show here, using inducible shRNA expression, that EKLF knockdown in mouse erythroleukemia (MEL) cells decreases erythrocytic and increases megakaryocytic as well as Fli-1 gene expression. Chromatin immunoprecipitation analyses revealed that the increase in megakaryocytic gene expression is associated with a marked increase in RNA pol II and FLI-1 occupancy at their promoters, albeit FLI-1 protein levels are only minimally affected. Similarly, we show that human CD34+ progenitors infected with shRNA lentivirus allowing EKLF knockdown generate an increased number of differentiated megakaryocytic cells associated with increased levels of megakaryocytic and Fli-1 gene transcripts. Single-cell progeny analysis of a cell population enriched in bipotent progenitors revealed that EKLF knockdown increases the number of megakaryocytic at the expense of erythrocytic colonies. Taken together, these data indicate that EKLF restricts megakaryocytic differentiation to the benefit of erythrocytic differentiation and suggest that this might be at least partially mediated by the inhibition of FLI-1 recruitment to megakaryocytic and Fli-1 gene promoters.
Collapse
|
21
|
Gordon AR, Outram SV, Keramatipour M, Goddard CA, Colledge WH, Metcalfe JC, Hager-Theodorides AL, Crompton T, Kemp PR. Splenomegaly and Modified Erythropoiesis in KLF13–/– Mice. J Biol Chem 2008; 283:11897-904. [DOI: 10.1074/jbc.m709569200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
22
|
Non-random subcellular distribution of variant EKLF in erythroid cells. Exp Cell Res 2008; 314:1595-604. [PMID: 18329016 DOI: 10.1016/j.yexcr.2008.01.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 01/11/2008] [Accepted: 01/29/2008] [Indexed: 11/22/2022]
Abstract
EKLF protein plays a prominent role during erythroid development as a nuclear transcription factor. Not surprisingly, exogenous EKLF quickly localizes to the nucleus. However, using two different assays we have unexpectedly found that a substantial proportion of endogenous EKLF resides in the cytoplasm at steady state in all erythroid cells examined. While EKLF localization does not appear to change during either erythroid development or terminal differentiation, we find that the protein displays subtle yet distinct biochemical and functional differences depending on which subcellular compartment it is isolated from, with PEST sequences possibly playing a role in these differences. Localization is unaffected by inhibition of CRM1 activity and the two populations are not differentiated by stability. Heterokaryon assays demonstrate that EKLF is able to shuttle out of the nucleus although its nuclear re-entry is rapid. These studies suggest there is an unexplored role for EKLF in the cytoplasm that is separate from its well-characterized nuclear function.
Collapse
|
23
|
Fuhrken PG, Chen C, Apostolidis PA, Wang M, Miller WM, Papoutsakis ET. Gene Ontology-driven transcriptional analysis of CD34+ cell-initiated megakaryocytic cultures identifies new transcriptional regulators of megakaryopoiesis. Physiol Genomics 2008; 33:159-69. [PMID: 18252802 DOI: 10.1152/physiolgenomics.00127.2007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Differentiation of hematopoietic stem and progenitor cells is an intricate process controlled in large part at the level of transcription. While some key megakaryocytic transcription factors have been identified, the complete network of megakaryocytic transcriptional control is poorly understood. Using global gene expression microarray analysis, Gene Ontology-based functional annotations, and a novel interlineage comparison with parallel, isogenic granulocytic cultures as a negative control, we closely examined the mRNA level of transcriptional regulators in megakaryocytes derived from human mobilized peripheral blood CD34(+) hematopoietic cells. This approach identified 199 differentially expressed transcription factors or transcriptional regulators. We identified and detailed the transcriptional kinetics of most known megakaryocytic transcription factors including GATA1, FLI1, and MAFG. Furthermore, many genes with transcription factor activity or transcription factor binding activity were identified in megakaryocytes that had not previously been associated with that lineage, including BTEB1, NR4A2, FOXO1A, MEF2C, HDAC5, VDR, and several genes associated with the tumor suppressor p53 (HIPK2, FHL2, and TADA3L). Protein expression and nuclear localization were confirmed in megakaryocytic cells for four of the novel candidate megakaryocytic transcription factors: FHL2, MXD1, E2F3, and RFX5. In light of the hypothesis that transcription factors expressed in a particular differentiation program are important contributors to such a program, these data substantially expand our understanding of transcriptional regulation in megakaryocytic differentiation of stem and progenitor cells.
Collapse
Affiliation(s)
- Peter G Fuhrken
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
| | | | | | | | | | | |
Collapse
|
24
|
Sumoylation of EKLF promotes transcriptional repression and is involved in inhibition of megakaryopoiesis. Mol Cell Biol 2007; 27:8547-60. [PMID: 17938210 DOI: 10.1128/mcb.00589-07] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Erythroid Krüppel-like factor (EKLF [KLF1]) is a transcriptional regulator that plays a critical role within a specific subset of hematopoietic cells, particularly in the erythroid lineage and its immediate precursor, the megakaryocyte-erythroid progenitor (MEP). We find that EKLF is posttranslationally modified by sumoylation at a single site near its amino terminus and that PIAS1 plays a critical role in this process. Mutation of this site has little effect on EKLF's ability to function as a transcriptional activator; however, it has a dramatic effect on its repressive abilities. The mechanism of repression likely involves a novel small ubiquitin-related modifier (SUMO)-dependent EKLF interaction with the Mi-2beta component of the NuRD repression complex. Mutated EKLF is attenuated in its ability to repress megakaryocyte differentiation, implicating EKLF sumoylation status in differentiative decisions emanating from the MEP. These studies demonstrate a novel mechanism by which transcription factor sumoylation can alter protein-protein interactions and bipotential lineage decisions.
Collapse
|
25
|
Bruce SJ, Gardiner BB, Burke LJ, Gongora MM, Grimmond SM, Perkins AC. Dynamic transcription programs during ES cell differentiation towards mesoderm in serum versus serum-freeBMP4 culture. BMC Genomics 2007; 8:365. [PMID: 17925037 PMCID: PMC2204012 DOI: 10.1186/1471-2164-8-365] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 10/10/2007] [Indexed: 12/30/2022] Open
Abstract
Background Expression profiling of embryonic stem (ES) cell differentiation in the presence of serum has been performed previously. It remains unclear if transcriptional activation is dependent on complex growth factor mixtures in serum or whether this process is intrinsic to ES cells once the stem cell program has been inactivated. The aims of this study were to determine the transcriptional programs associated with the stem cell state and to characterize mesoderm differentiation between serum and serum-free culture. Results ES cells were differentiated as embryoid bodies in 10% FBS or serum-free media containing BMP4 (2 ng/ml), and expression profiled using 47 K Illumina(R) Sentrix arrays. Statistical methods were employed to define gene sets characteristic of stem cell, epiblast and primitive streak programs. Although the initial differentiation profile was similar between the two culture conditions, cardiac gene expression was inhibited in serum whereas blood gene expression was enhanced. Also, expression of many members of the Kruppel-like factor (KLF) family of transcription factors changed dramatically during the first few days of differentiation. KLF2 and KLF4 co-localized with OCT4 in a sub-nuclear compartment of ES cells, dynamic changes in KLF-DNA binding activities occurred upon differentiation, and strong bio-informatic evidence for direct regulation of many stem cell genes by KLFs was found. Conclusion Down regulation of stem cell genes and activation of epiblast/primitive streak genes is similar in serum and defined media, but subsequent mesoderm differentiation is strongly influenced by the composition of the media. In addition, KLF family members are likely to be important regulators of many stem cell genes.
Collapse
Affiliation(s)
- Stephen J Bruce
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
Gardiner MR, Gongora MM, Grimmond SM, Perkins AC. A global role for zebrafish klf4 in embryonic erythropoiesis. Mech Dev 2007; 124:762-74. [PMID: 17709232 DOI: 10.1016/j.mod.2007.06.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 06/06/2007] [Accepted: 06/18/2007] [Indexed: 01/21/2023]
Abstract
There are two waves of erythropoiesis, known as primitive and definitive waves in mammals and lower vertebrates including zebrafish. The founding member of the Kruppel-like factor (KLF) family of CACCC-box binding proteins, EKLF/Klf1, is essential for definitive erythropoiesis in mammals but only plays a minor role in primitive erythropoiesis. Morpholino knockdown experiments have shown a role for zebrafish klf4 in primitive erythropoiesis and hatching gland formation. In order to generate a global understanding of how klf4 might influence gene expression and differentiation, we have performed expression profiling of klf4 morphants, and then performed validation of many putative target genes by qRT-PCR and whole mount in situ hybridization. We found a critical role for klf4 in embryonic globin, heme synthesis and hatching gland gene expression. In contrast, there was an increase in expression of definitive hematopoietic specific genes such as larval globin genes, runx1 and c-myb from 24 hpf, suggesting a selective role for klf4 in primitive rather than definitive erythropoiesis. In addition, we show klf4 preferentially binds CACCC box elements in the primitive zebrafish beta-like globin gene promoters. These results have global implications for primitive erythroid gene regulation by KLF-CACCC box interactions.
Collapse
Affiliation(s)
- M R Gardiner
- Institute for Molecular Bioscience, University of Queensland, Australia
| | | | | | | |
Collapse
|
27
|
Frontelo P, Manwani D, Galdass M, Karsunky H, Lohmann F, Gallagher PG, Bieker JJ. Novel role for EKLF in megakaryocyte lineage commitment. Blood 2007; 110:3871-80. [PMID: 17715392 PMCID: PMC2190608 DOI: 10.1182/blood-2007-03-082065] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Megakaryocytes and erythroid cells are thought to derive from a common progenitor during hematopoietic differentiation. Although a number of transcriptional regulators are important for this process, they do not explain the bipotential result. We now show by gain- and loss-of-function studies that erythroid Krüppel-like factor (EKLF), a transcription factor whose role in erythroid gene regulation is well established, plays an unexpected directive role in the megakaryocyte lineage. EKLF inhibits the formation of megakaryocytes while at the same time stimulating erythroid differentiation. Quantitative examination of expression during hematopoiesis shows that, unlike genes whose presence is required for establishment of both lineages, EKLF is uniquely down-regulated in megakaryocytes after formation of the megakaryocyte-erythroid progenitor. Expression profiling and molecular analyses support these observations and suggest that megakaryocytic inhibition is achieved, at least in part, by EKLF repression of Fli-1 message levels.
Collapse
|
28
|
Tallack MR, Keys JR, Perkins AC. Erythroid Kruppel-like Factor Regulates the G1 Cyclin Dependent Kinase Inhibitor p18INK4c. J Mol Biol 2007; 369:313-21. [PMID: 17442339 DOI: 10.1016/j.jmb.2007.02.109] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 02/25/2007] [Accepted: 02/28/2007] [Indexed: 12/18/2022]
Abstract
Erythroid Kruppel-like factor (EKLF, KLF1) is an essential erythroid cell specific C(2)H(2) zinc finger transcription factor that binds CACC box elements in promoters and distant regulatory elements to activate transcription. Forced expression of EKLF arrests cell division. The cyclin dependent kinase (Cdk) inhibitor p18(INK4c) was identified as a potential novel EKLF target gene from an expression profiling study. The p18(INK4c) protein functions as an inhibitor of Cdk4 and Cdk6 activity during early G1 phase of the cell cycle, thus acting as a physiological brake on cell division. We confirmed p18(INK4c) is downregulated in EKLF null mice by real-time PCR and Western blotting, and identified three closely associated and highly conserved EKLF binding sites (CCNCNCCCN) approximately 1 kb upstream of the p18(INK4c) transcriptional start site. We showed that EKLF binds to one of these elements by gel shift assay and demonstrated this site is capable of driving EKLF dependent transcription. We also determined by chromatin immunoprecipitation (ChIP) that this region of the p18(INK4c) promoter is bound by EKLF in erythroid cells. Thus, EKLF is a direct regulator of p18(INK4c) gene expression, and much of EKLF's role in driving erythroid cell differentiation may occur via p18(INK4c).
Collapse
Affiliation(s)
- Michael R Tallack
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, 4072, Australia
| | | | | |
Collapse
|
29
|
Keys JR, Tallack MR, Hodge DJ, Cridland SO, David R, Perkins AC. Genomic organisation and regulation of murine alpha haemoglobin stabilising protein by erythroid Kruppel-like factor. Br J Haematol 2006; 136:150-7. [PMID: 17069580 DOI: 10.1111/j.1365-2141.2006.06381.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alpha haemoglobin stabilising protein (AHSP) binds free alpha-globin chains and plays an important role in the protection of red cells, particularly during beta-thalassaemia. Murine ASHP was discovered as a GATA-1 target gene and human AHSP is directly regulated by GATA-1. More recently, AHSP was rediscovered as a highly erythroid Kruppel-like factor (EKLF) -dependent transcript. We have determined the organisation of the murine AHSP gene and compared it to orthologs. There are two CACC box elements in the proximal promoter. The proximal element is absolutely conserved, but does not bind EKLF as it is not a canonical binding site. In rodents, the distal element contains a 3 bp insertion that disrupts the typical EKLF binding consensus region. Nevertheless, EKLF binds this atypical site by gel mobility shift assay, specifically occupies the AHSP promoter in vivo in a chromatin immunoprecipitation assay, and transactivates AHSP through this CACC site in promoter-reporter assays. Our results suggest EKLF can occupy CACC elements in vivo that are not predictable from the consensus binding site inferred from structural studies. We also propose that absence of AHSP in EKLF-null red cells exacerbates the toxicity of free alpha-globin chains, which exist because of the defect in beta-globin gene activation.
Collapse
Affiliation(s)
- Janelle R Keys
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Qld, Australia
| | | | | | | | | | | |
Collapse
|
30
|
Nilson DG, Sabatino DE, Bodine DM, Gallagher PG. Major erythrocyte membrane protein genes in EKLF-deficient mice. Exp Hematol 2006; 34:705-12. [PMID: 16728274 DOI: 10.1016/j.exphem.2006.02.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 02/16/2006] [Accepted: 02/21/2006] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Mice deficient in the transcription factor erythroid Krüppel-like factor, KLF1 (EKLF) die approximately 14.5 days postcoitum of anemia, attributed to decreased expression of the beta-globin gene. The objectives of this study were to rescue EKLF-deficient embryos with mice expressing gamma-globin from beta-spectrin or ankyrin promoters and to characterize expression of the major erythrocyte membrane genes in EKLF-deficient cells. METHODS Transgenic beta-spectrin/gamma-globin or ankyrin/gamma-globin mice were bred onto EKLF-deficient and wild-type backgrounds. Animals were genotyped, gamma-globin mRNA levels measured, and hemoglobin electrophoresis performed. Steady-state mRNA levels and transcriptional rates of the major erythrocyte membrane protein genes were assayed. RESULTS beta-spectrin/gamma-globin or ankyrin/gamma-globin mice on EKLF-deficient and wild-type backgrounds had identical levels of gamma-globin mRNA, indicating EKLF-independence of these promoters. gamma-Globin expression improved globin chain imbalance, but hemolysis was not improved and no live-born EKLF-deficient/(A)gamma-globin mice were obtained. Circulating erythroid cells from EKLF-deficient/(A)gamma-globin embryos exhibited hemolysis reminiscent of that seen in patients with severe erythrocyte membrane defects. Levels of beta-spectrin, ankyrin, and band 3 mRNA, but not alpha-spectrin, were decreased in EKLF-deficient fetal liver RNA. In a run-on assay, levels of transcription of the ankyrin and band 3 genes were decreased in EKLF-deficient fetal liver nuclei. CONCLUSIONS These results indicate that the EKLF-responsive regions of the ankyrin and beta-spectrin genes are outside their promoters and that EKLF is necessary for full transcriptional activity of the ankyrin and band 3 genes; the results also provide additional evidence that defects in addition to beta-globin deficiency, including an abnormal erythrocyte membrane, contribute to the anemia and embryonic lethality in EKLF-deficient mice.
Collapse
Affiliation(s)
- Douglas G Nilson
- Hematopoiesis Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
31
|
Pilon AM, Nilson DG, Zhou D, Sangerman J, Townes TM, Bodine DM, Gallagher PG. Alterations in expression and chromatin configuration of the alpha hemoglobin-stabilizing protein gene in erythroid Kruppel-like factor-deficient mice. Mol Cell Biol 2006; 26:4368-77. [PMID: 16705186 PMCID: PMC1489081 DOI: 10.1128/mcb.02216-05] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Erythroid Krüppel-like factor (EKLF) is an erythroid zinc finger protein identified by its interaction with a CACCC sequence in the beta-globin promoter, where it establishes local chromatin structure permitting beta-globin gene transcription. We sought to identify other EKLF target genes and determine the chromatin status of these genes in the presence and absence of EKLF. We identified alpha hemoglobin-stabilizing protein (AHSP) by subtractive hybridization and demonstrated a 95 to 99.9% reduction in AHSP mRNA and the absence of AHSP in EKLF-deficient cells. Chromatin at the AHSP promoter from EKLF-deficient cells lacked a DNase I hypersensitive site and exhibited histone hypoacetylation across the locus compared to hyperacetylation of wild-type chromatin. Wild-type chromatin demonstrated a peak of EKLF binding over a promoter region CACCC box that differs from the EKLF consensus by a nucleotide. In mobility shift assays, the AHSP promoter CACCC site bound EKLF in a manner comparable to the beta-globin promoter CACCC site, indicating a broader recognition sequence for the EKLF consensus binding site. The AHSP promoter was transactivated by EKLF in K562 cells, which lack EKLF. These results support the hypothesis that EKLF acts as a transcription factor and a chromatin modulator for the AHSP and beta-globin genes and indicate that EKLF may play similar roles for other erythroid genes.
Collapse
Affiliation(s)
- Andre M Pilon
- Hematopoiesis Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-4442, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Wein MN, Jones DC, Glimcher LH. Turning down the system: counter-regulatory mechanisms in bone and adaptive immunity. Immunol Rev 2005; 208:66-79. [PMID: 16313341 DOI: 10.1111/j.0105-2896.2005.00322.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Major advances have been made in recent years toward the identification of transcription factors that control cell-type-specific gene expression in the skeletal and adaptive immune systems. However, the identification of factors necessary and sufficient to drive production of effector cell proteins such as matrix components and cytokines represents the first step toward understanding how cells in bone and the adaptive system achieve their highly specialized functions. Here, we provide selected examples of counter-regulatory mechanisms that serve to turn down cells involved in extracellular matrix biosynthesis and adaptive immunity at the level of the transcription factors Runx2 and nuclear factor for the activation of T cells.
Collapse
Affiliation(s)
- Marc N Wein
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | |
Collapse
|
33
|
Huo X, Zhang J. Important roles of reversible acetylation in the function of hematopoietic transcription factors. J Cell Mol Med 2005; 9:103-12. [PMID: 15784168 PMCID: PMC6741356 DOI: 10.1111/j.1582-4934.2005.tb00340.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Hematopoiesis is a very complex process whose proper functioning requires the regulated action of a number of transcription factors. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) play significant roles in the regulation of hematopoietic transcription factors activity. Transcription factors such as GATA-1, EKLF, NF-E2, GATA-1, PU.1 recruit HATs and HDACs to chromatin, leading to histone acetylation and deacetylation, that affect chromatin structure and result in gene expression changes. On the other hand, transcription factors themselves can be acetylated and deacetylated by HATs and HDACs, respectively. Consequently, some important functions of these transcription factors are influenced, including DNA binding, transcription activation, repressor activity and proteinprotein interactions. The regulation of hematopoietic transcription factors activity by HATs and HDACs may serve as a good model for studying how tissue-specific and lineage-specific gene expression is controlled through acetylation/ deacetylation of histone/nonhistone proteins.
Collapse
Affiliation(s)
- Xiaofang Huo
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, The Chinese Academy of Medical Sciences and Peking Union Medical College, Dong Dan San Tiao 5, Beijing 100005, China
| | | |
Collapse
|
34
|
Abstract
Dynamic changes in transcription factor function can be mediated by switching its interaction with coactivators and corepressors. Erythroid Kruppel-like factor (EKLF) is an erythroid cell-specific transcription factor that plays a critical role in beta-globin gene activation via its interactions with CBP/p300 and SWI/SNF proteins. Unexpectedly, it also interacts with Sin3A and histone deacetylase 1 (HDAC1) corepressors via its zinc finger domain. We now find that selected point mutants can uncouple activation and repression and that an intact finger structure is not required for interactions with Sin3A/HDAC1 or for transrepression. Most intriguingly, EKLF repression exhibits stage specificity, with reversible EKLF-Sin3A interactions playing a key role in this process. Finally, we have located a key lysine residue that is both a substrate for CBP acetylation and required for Sin3A interaction. These data suggest a model whereby the stage of the erythroid cell alters the acetylation status of EKLF and plays a critical role in directing its coactivator-corepressor interactions and downstream transcriptional effects.
Collapse
Affiliation(s)
- Xiaoyong Chen
- Mount Sinai School of Medicine, Brookdale Department of Molecular, Cell and Developmental Biology, Box 1020, One Gustave L. Levy Place, New York, NY 10029, USA
| | | |
Collapse
|
35
|
Drissen R, Palstra RJ, Gillemans N, Splinter E, Grosveld F, Philipsen S, de Laat W. The active spatial organization of the beta-globin locus requires the transcription factor EKLF. Genes Dev 2004; 18:2485-90. [PMID: 15489291 PMCID: PMC529536 DOI: 10.1101/gad.317004] [Citation(s) in RCA: 285] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Three-dimensional organization of a gene locus is important for its regulation, as recently demonstrated for the beta-globin locus. When actively expressed, the cis-regulatory elements of the beta-globin locus are in proximity in the nuclear space, forming a compartment termed the Active Chromatin Hub (ACH). However, it is unknown which proteins are involved in ACH formation. Here, we show that EKLF, an erythroid transcription factor required for adult beta-globin gene transcription, is also required for ACH formation. We conclude that transcription factors can play an essential role in the three-dimensional organization of gene loci.
Collapse
Affiliation(s)
- Roy Drissen
- Erasmus University Medical Center, Department of Cell Biology, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
36
|
Xue L, Chen X, Chang Y, Bieker JJ. Regulatory elements of the EKLF gene that direct erythroid cell-specific expression during mammalian development. Blood 2004; 103:4078-83. [PMID: 14764531 DOI: 10.1182/blood-2003-09-3231] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AbstractErythroid Krüppel-like factor (EKLF) plays an essential role in enabling β-globin expression during erythroid ontogeny. It is first expressed in the extraembryonic mesoderm of the yolk sac within the morphologically unique cells that give rise to the blood islands, and then later within the hepatic primordia. The BMP4/Smad pathway plays a critical role in the induction of EKLF, and transient transfection analyses demonstrate that sequences located within less than 1 kb of its transcription initiation site are sufficient for high-level erythroid-specific transcription. We have used transgenic analyses to verify that 950 bp located adjacent to the EKLF start site of transcription is sufficient to generate lacZ expression within the blood islands as well as the fetal liver during embryonic development. Of particular importance are 3 regions, 2 of which overlap endogenous erythroid-specific DNase hypersensitive sites, and 1 of which includes the proximal promoter region. The onset of transgene expression mimics that of endogenous EKLF as it begins by day 7.5 (d7.5) to d8.0. In addition, it exhibits a strict hematopoietic specificity, localized only to these cells and not to the adjacent vasculature at all stages examined. Finally, expression is heterocellular, implying that although these elements are sufficient for tissue-specific expression, they do not shield against the position effects of adjacent chromatin. These analyses demonstrate that a surprisingly small DNA segment contains all the information needed to target a linked gene to the hematopoietic compartment at both early and later stages of development, and may be a useful cassette for this purpose.
Collapse
Affiliation(s)
- Li Xue
- Mount Sinai School of Medicine, Brookdale Department of Molecular, Cell, and Developmental Biology, Box 1020, One Gustave L. Levy Pl, New York, NY 10029, USA
| | | | | | | |
Collapse
|
37
|
Dzikaite V, Hultcrantz R, Melefors O. The regulatory effect of heme on erythroid aminolevulinate synthase in natural erythroid cells. ACTA ACUST UNITED AC 2003; 1630:19-24. [PMID: 14580675 DOI: 10.1016/j.bbaexp.2003.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A major enzymatic pathway in erythroid cells is the eight-step formation of heme, starting with the erythroid isoform of aminolevulinate synthase (eALAS). We studied the regulation of eALAS synthesis by heme in natural erythroid cells. Erythroid cells from mouse blood or bone marrow were incubated with different concentrations of heme and labelled with [35S]methionine. This was followed by immunoprecipitation of eALAS proteins. Northern blot analysis was done on mRNA isolated from bone marrow. Incubation with heme (5-100 muM) was shown to clearly inhibit eALAS synthesis in erythroid cells of bone marrow. This inhibitory effect of heme could also be observed in peripheral blood cells at higher concentrations while the preform of eALAS was rather increased. However, at lower concentrations of heme (1-10 microM), eALAS synthesis increased. Northern blot studies argued the inhibitory effect was at the posttranscriptional level. Our results suggest that the net effect of murine eALAS regulation by heme varies with the degree of erythroid differentiation. Heme formation seems to be more tightly controlled in the bone marrow (nucleated) cells in order to prevent oxidative cell damage, compared to more differentiated erythroid cells.
Collapse
Affiliation(s)
- Vijole Dzikaite
- Department of Gastroenterology and Hepatology, Karolinska Hospital, Stockholm SE-171 76, Sweden.
| | | | | |
Collapse
|
38
|
Zhao J, Bian ZC, Yee K, Chen BPC, Chien S, Guan JL. Identification of transcription factor KLF8 as a downstream target of focal adhesion kinase in its regulation of cyclin D1 and cell cycle progression. Mol Cell 2003; 11:1503-15. [PMID: 12820964 DOI: 10.1016/s1097-2765(03)00179-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Focal adhesion kinase (FAK) is an important mediator of integrin signaling in the regulation of cell adhesion, migration, survival, and proliferation. Here we report the identification of the transcription factor KLF8 as a target of FAK in cell cycle regulation. KLF8 is induced by FAK and decreased by FAK dominant-negative mutant DeltaC14. Overexpression of KLF8 increases cell cycle progression, whereas inhibition of endogenous KLF8 by siRNA reduces it. Cyclin D1 promoter is identified as a target of KLF8, which is activated both directly by KLF8 binding to the GT box A and by an indirect mechanism through its repression of a potential inhibitory regulator of cyclin D1. Transcription activation of cyclin D1 by FAK requires both Ets family and KLF8 factors in a temporally differential manner. Together, our data provide further insights into molecular mechanism for FAK to regulate cell cycle progression.
Collapse
Affiliation(s)
- Jihe Zhao
- Department of Molecular Medicine, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | | | | | | | | | | |
Collapse
|
39
|
Chrisman HR, Tindall DJ. Identification and characterization of a consensus DNA binding element for the zinc finger transcription factor TIEG/EGRalpha. DNA Cell Biol 2003; 22:187-99. [PMID: 12804117 DOI: 10.1089/104454903321655819] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
TGFbeta-Inducible Early Gene (TIEG) and the alternatively-transcribed Early Growth Response Gene alpha (EGRalpha) share a Cys(2)His(2) three-zinc finger region with high homology to Sp1 within its zinc finger region. Three-zinc finger transcription factors bind to GC-rich sequences, with small variations in consensus sequence between subfamilies. In this work, a consensus sequence was identified for TIEG/EGRalpha by expressing and purifying the zinc finger region of the protein, and using this to select a binding site from a random oligonucleotide library by iterative cycles of nitrocellulose filter binding and PCR. A fusion of the TIEG/EGRalpha with the VP16 activation domain supported transcription from this site when cloned in front of a heterologous promoter. Mutational analysis of the binding site identified a GT-rich core (5'-GGTGTG-3') that was necessary for binding, with mutations outside of this region causing only a small to moderate decrease in binding.
Collapse
Affiliation(s)
- Holly R Chrisman
- Department of Urology Research, Mayo Clinic/Foundation, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
40
|
Jackson DA, McDowell JC, Dean A. Beta-globin locus control region HS2 and HS3 interact structurally and functionally. Nucleic Acids Res 2003; 31:1180-90. [PMID: 12582237 PMCID: PMC150235 DOI: 10.1093/nar/gkg217] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The overall structure of the DNase I hypersensitive sites (HSs) that comprise the beta-globin locus control region (LCR) is highly conserved among mammals, implying that the HSs have conserved functions. However, it is not well understood how the LCR HSs, either individually or collectively, activate transcription. We analyzed the interactions of HS2, HS3 and HS4 with the human epsilon- and beta-globin genes in chromatinized episomes in fetal/embryonic K562 cells. Only HS2 activates transcription of the epsilon-globin gene, while all three HSs activate the beta-globin gene. HS3 stimulates the beta-globin gene constitutively, but HS2 and HS4 transactivation requires expression of the transcription factor EKLF, which is not present in K562 cells but is required for beta-globin expression in vivo. To begin addressing how the individual HSs may interact with one another in a complex, we linked the beta-globin gene to both the HS2 and HS3. HS2 and HS3 together resulted in synergistic stimulation of beta-globin transcription. Unexpectedly, mutated, inactive forms of HS2 impeded the activation of the beta-globin gene by HS3. Thus, there appear to be distinct interactions among the HSs and between the HSs and the globin genes. These preferential, non-exclusive interactions may underlie an important structural and functional cooperativity among the regulatory sequences of the beta-globin locus in vivo.
Collapse
Affiliation(s)
- David A Jackson
- Laboratory of Cellular and Developmental Biology, NIDDK, NIH, Bethesda, MD 20892-2715, USA
| | | | | |
Collapse
|
41
|
Irenge LM, Heusterspreute M, Philippe M, Derclaye I, Robert A, Gala JL. Validation of a Recombinant DNA Construct (μLCR and Full-Length β-Globin Gene) for Quantification of Human β-Globin Expression: Application to Mutations in the Promoter, Intronic, and 5′- and 3′-Untranslated Regions of the Human β-Globin Gene. Clin Chem 2002. [DOI: 10.1093/clinchem/48.10.1787] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Leonid M Irenge
- Applied Molecular Technologies, Center for Human Genetics, Université Catholique de Louvain, Clos-Chapelle-aux-Champs, 30-UCL/30.46, B-1200 Bruxelles, Belgium
| | - Michel Heusterspreute
- Applied Molecular Technologies, Center for Human Genetics, Université Catholique de Louvain, Clos-Chapelle-aux-Champs, 30-UCL/30.46, B-1200 Bruxelles, Belgium
| | - Marianne Philippe
- Department of Biochemistry, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Avenue Hippocrate, 30, B-1200 Bruxelles, Belgium
| | - Isabelle Derclaye
- Applied Molecular Technologies, Center for Human Genetics, Université Catholique de Louvain, Clos-Chapelle-aux-Champs, 30-UCL/30.46, B-1200 Bruxelles, Belgium
| | - Annie Robert
- Biostatistics and Epidemiology, Clos-Chapelle-aux-Champs, 30-UCL/30.34, Université Catholique de Louvain, B-1200 Bruxelles, Belgium
| | - Jean-Luc Gala
- Applied Molecular Technologies, Center for Human Genetics, Université Catholique de Louvain, Clos-Chapelle-aux-Champs, 30-UCL/30.46, B-1200 Bruxelles, Belgium
- Applied Molecular Technologies, Queen Astrid Military Hospital, Rue Bruyn, 2, B-1120 Bruxelles, Belgium
| |
Collapse
|
42
|
Perry C, Soreq H. Transcriptional regulation of erythropoiesis. Fine tuning of combinatorial multi-domain elements. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:3607-18. [PMID: 12153557 DOI: 10.1046/j.1432-1033.2002.02999.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Haematopoiesis, the differentiation of haematopoietic stem cells and progenitors into various lineages, involves complex interactions of transcription factors that modulate the expression of downstream genes and mediate proliferation and differentiation signals. Commitment of pluripotent haematopoietic stem cells to the erythroid lineage induces erythropoiesis, the production of red blood cells. This process involves a concerted progression through an erythroid burst forming unit (BFU-E), an erythroid colony forming unit (CFU-E), proerythroblast and an erythroblast. The terminally differentiated erythrocytes, in mammals, lose their nucleus yet function several more months. A well-coordinated cohort of transcription factors regulates the formation, survival, proliferation and differentiation of multipotent progenitor into the erythroid lineage. Here, we discuss broad-spectrum factors essential for self-renewal and/or differentiation of multipotent cells as well as specific factors required for proper erythroid development. These factors may operate solely or as part of transcriptional complexes, and exert activation or repression. Sequence comparisons reveal evolutionarily conserved modular composition for these factors; X-ray crystallography demonstrates that they include multidomain elements (e.g. HLH or zinc finger motifs), consistent with their complex interactions with other proteins. Finally, transfections and genomic studies show that the timing of each factor's expression during the hematopoietic process, the cell lineages affected and the existing combination of other factors determine the erythroid cell fate.
Collapse
Affiliation(s)
- Chava Perry
- Department of Biological Chemistry, The Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | | |
Collapse
|
43
|
Pandya K, Townes TM. Basic residues within the Kruppel zinc finger DNA binding domains are the critical nuclear localization determinants of EKLF/KLF-1. J Biol Chem 2002; 277:16304-12. [PMID: 11844803 DOI: 10.1074/jbc.m200866200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
EKLF/KLF-1 is an erythroid-restricted transcription factor essential for expression of the adult beta-globin gene. EKLF/KLF-1 is a 358-amino acid nuclear protein with an amino-terminal proline-rich domain and a carboxyl-terminal DNA binding domain. The nuclear localization signal (NLS) of EKLF/KLF-1 has not been empirically determined. We generated a series of epitope-tagged deletion and point mutants and assessed their subcellular localization. Our results delimit the NLS to the 83-amino acid (amino acids 276-358) DNA binding domain that consists of three Kruppel zinc fingers. All three zinc fingers are necessary for efficient nuclear localization; deletion of any individual finger results in cytoplasmic accumulation. Fusion of the three zinc fingers to green fluorescent protein (GFP) targeted GFP to the nucleus, demonstrating that the zinc finger domain is sufficient for nuclear localization. EKLF/KLF-1 containing histidine to alanine mutations that disrupt the structure of all three fingers retains appropriate nuclear localization, indicating that neither the tertiary structure of the zinc fingers nor specific DNA binding are necessary for nuclear localization. We demonstrate that basic residues within the fingers are the critical determinants for nuclear localization; mutations of these basic residues to alanine resulted in cytoplasmic mislocalization. The basic residues of all mammalian Kruppel zinc fingers are highly conserved; therefore we propose that these basic residues are a common NLS shared by all Kruppel family members.
Collapse
Affiliation(s)
- Kumar Pandya
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
44
|
Adelman CA, Chattopadhyay S, Bieker JJ. The BMP/BMPR/Smad pathway directs expression of the erythroid-specific EKLF and GATA1 transcription factors during embryoid body differentiation in serum-free media. Development 2002; 129:539-49. [PMID: 11807044 DOI: 10.1242/dev.129.2.539] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Erythroid cell-specific gene regulation during terminal differentiation is controlled by transcriptional regulators, such as EKLF and GATA1, that themselves exhibit tissue-restricted expression patterns. Their early expression, already in evidence within multipotential hematopoietic cell lines, has made it difficult to determine what extracellular effectors and transduction mechanisms might be directing the onset of their own transcription during embryogenesis. To circumvent this problem, we have taken the novel approach of investigating whether the ability of embryonic stem (ES) cells to mimic early developmental patterns of cellular expression during embryoid body (EB) differentiation can address this issue. We first established conditions whereby EBs could form efficiently in the absence of serum. Surprisingly, in addition to mesoderm, these cells expressed hemangioblast and hematopoietic markers. However, they did not express the committed erythroid markers EKLF and GATA1, nor the terminally differentiated β-like globin markers. Using this system, we determined that EB differentiation in BMP4 was necessary and sufficient to recover EKLF and GATA1 expression and could be further stimulated by the inclusion of VEGF, SCF, erythropoietin and thyroid hormone. EBs were competent to respond to BMP4 only until day 4 of differentiation, which coincides with the normal onset of EKLF expression. The direct involvement of the BMP/Smad pathway in this induction process was further verified by showing that erythroid expression of a dominant negative BMP1B receptor or of the inhibitory Smad6 protein prevented induction of EKLF or GATA1 even in the presence of serum. Although Smad1, Smad5 and Smad8 are all expressed in the EBs, BMP4 induction of EKLF and GATA1 transcription is not immediate. These data implicate the BMP/Smad induction system as being a crucial pathway to direct the onset of EKLF and GATA1 expression during hematopoietic differentiation and demonstrate that EB differentiation can be manipulated to study induction of specific genes that are expressed early within a lineage.
Collapse
Affiliation(s)
- Carrie A Adelman
- Department of Biochemistry and Molecular Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
45
|
Mahajan MC, Weissman SM. DNA-dependent adenosine triphosphatase (helicaselike transcription factor) activates beta-globin transcription in K562 cells. Blood 2002; 99:348-56. [PMID: 11756191 DOI: 10.1182/blood.v99.1.348] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Correct developmental regulation of beta-like globin gene expression is achieved by preferential transcription of a gene at a given developmental stage, silencing of other beta-like gene promoters, and competition among these promoters for interaction with the locus control region (LCR). Several evolutionarily conserved DNA elements in the promoters of the beta-like genes and LCR have been studied in detail, and the role of their binding factors has been investigated. However, the beta-globin promoter includes additional evolutionarily conserved sequences of unknown function. The present study examined the properties of a 21-base pair (bp) promoter-conserved sequence (PCS) located at positions -115 to -136 bp relative to the transcription start site of the beta-globin gene. A helicaselike transcription factor (HLTF) belonging to the SWI2/SNF2 family of proteins binds to the PCS and a partly homologous sequence in the enhancer region of the LCR hypersensitive site 2 (HS2). Elevation of the level of HLTF in K562 erythroleukemic cells increases beta-promoter activity in transient transfection experiments, and mutations in the PCS that remove HLTF-binding regions abolish this effect, suggesting that HLTF is an activator of beta-globin transcription. Overexpression of HLTF in K562 cells does not affect the endogenous levels of gamma- and epsilon-globin message, but it markedly activates beta-globin transcription. In conclusion, this study reports a transcription factor belonging to the SWI2/SNF2 family, which preferentially activates chromosomal beta-globin gene transcription and which has not previously been implicated in globin gene regulation.
Collapse
Affiliation(s)
- Milind C Mahajan
- Department of Genetics, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, CT 06536, USA
| | | |
Collapse
|
46
|
Sagasser M, Lu GH, Hahlbrock K, Weisshaar B. A. thaliana TRANSPARENT TESTA 1 is involved in seed coat development and defines the WIP subfamily of plant zinc finger proteins. Genes Dev 2002; 16:138-49. [PMID: 11782451 PMCID: PMC155310 DOI: 10.1101/gad.212702] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2001] [Accepted: 11/05/2001] [Indexed: 01/28/2023]
Abstract
Seeds of the Arabidopsis thaliana transparent testa 1 mutant (tt1) appear yellow, due to the lack of condensed tannin pigments in the seed coat. The TT1 gene was isolated by reverse genetics using an En-1 transposon mutagenized A. thaliana population. TT1 gene expression was detected in developing ovules and young seeds only, and the gene was shown to encode a nuclear protein. Mutant seeds displayed altered morphology of the seed endothelium in which brown tannin pigments accumulate in wild-type plants, indicating that TT1 is involved in the differentiation of this cell layer. When overexpressed in transgenic A. thaliana plants, TT1 caused aberrant development and organ morphology. The protein contains a novel combination of two TFIIIA-type zinc finger motifs. Closely related motifs were detected in a number of putative proteins deduced from plant genomic and EST sequences. The new protein domain containing this type of zinc finger motifs was designated WIP, according to three strictly conserved amino acid residues. Our data indicate the existence of a small gene family in A. thaliana which is defined by the occurrence of the WIP domain. WIP genes may play important roles in regulating developmental processes, including the control of endothelium differentiation.
Collapse
Affiliation(s)
- Martin Sagasser
- Max-Planck-Institut für Züchtungsforschung, Abteilung Biochemie, D-50829 Köln, Germany
| | | | | | | |
Collapse
|
47
|
Kaczynski J, Zhang JS, Ellenrieder V, Conley A, Duenes T, Kester H, van Der Burg B, Urrutia R. The Sp1-like protein BTEB3 inhibits transcription via the basic transcription element box by interacting with mSin3A and HDAC-1 co-repressors and competing with Sp1. J Biol Chem 2001; 276:36749-56. [PMID: 11477107 DOI: 10.1074/jbc.m105831200] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sp1-like proteins are characterized by three conserved C-terminal zinc finger motifs that bind GC-rich sequences found in promoters of numerous genes essential for mammalian cell homeostasis. These proteins behave as transcriptional activators or repressors. Although significant information has been reported on the molecular mechanisms by which Sp1-like activators function, relatively little is known about mechanisms for repressor proteins. Here we report the functional characterization of BTEB3, a ubiquitously expressed Sp1-like transcriptional repressor. GAL4 assays show that the N terminus of BTEB3 contains regions that can act as direct repressor domains. Immunoprecipitation assays reveal that BTEB3 interacts with the co-repressor mSin3A and the histone deacetylase protein HDAC-1. Gel shift assays demonstrate that BTEB3 specifically binds the BTE site, a well characterized GC-rich DNA element, with an affinity similar to that of Sp1. Reporter and gel shift assays in Chinese hamster ovary cells show that BTEB3 can also mediate repression by competing with Sp1 for BTE binding. Thus, the characterization of this protein expands the repertoire of BTEB-like members of the Sp1 family involved in transcriptional repression. Furthermore, our results suggest a mechanism of repression for BTEB3 involving direct repression by the N terminus via interaction with mSin3A and HDAC-1 and competition with Sp1 via the DNA-binding domain.
Collapse
Affiliation(s)
- J Kaczynski
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota 55901, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- J J Bieker
- Department of Biochemistry and Molecular Biology, Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
49
|
Zhang JS, Moncrieffe MC, Kaczynski J, Ellenrieder V, Prendergast FG, Urrutia R. A conserved alpha-helical motif mediates the interaction of Sp1-like transcriptional repressors with the corepressor mSin3A. Mol Cell Biol 2001; 21:5041-9. [PMID: 11438660 PMCID: PMC87230 DOI: 10.1128/mcb.21.15.5041-5049.2001] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sp1-like proteins are defined by three highly homologous C(2)H(2) zinc finger motifs that bind GC-rich sequences found in the promoters of a large number of genes essential for mammalian cell homeostasis. Here we report that TIEG2, a transforming growth factor beta-inducible Sp1-like protein with antiproliferative functions, represses transcription through recruitment of the mSin3A-histone deacetylase complex. The interaction of TIEG2 with mSin3A is mediated by an alpha-helical repression motif (alpha-HRM) located within the repression domain (R1) of TIEG2. This alpha-HRM specifically associates with the second paired amphipathic helix (PAH2) domain of mSin3A. Mutations in the TIEG2 alpha-HRM domain that disrupt its helical structure abolish its ability to both bind mSin3A and repress transcription. Interestingly, the alpha-HRM is conserved in both the TIEG (TIEG1 and TIEG2) and BTEB (BTEB1, BTEB3, and BTEB4) subfamilies of Sp1-like proteins. The alpha-HRM from these proteins also mediates direct interaction with mSin3A and represses transcription. Surprisingly, we found that the alpha-HRM of the Sp1-like proteins characterized here exhibits structural and functional resemblance to the Sin3A-interacting domain previously described for the basic helix-loop-helix protein Mad1. Thus, our study defines a mechanism of transcriptional repression via the interactions of the alpha-HRM with the Sin3-histone deacetylase complex that is utilized by at least five Sp1-like transcriptional factors. More importantly, we demonstrate that a helical repression motif which mediates Sin3 interaction is not an exclusive structural and functional characteristic of the Mad1 subfamily but rather has a wider functional impact on transcriptional repression than previously demonstrated.
Collapse
Affiliation(s)
- J S Zhang
- Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota 55901, USA
| | | | | | | | | | | |
Collapse
|
50
|
Buckley AF, Kuo CT, Leiden JM. Transcription factor LKLF is sufficient to program T cell quiescence via a c-Myc-dependent pathway. Nat Immunol 2001; 2:698-704. [PMID: 11477405 DOI: 10.1038/90633] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
T lymphocytes circulate in a quiescent state until they encounter cognate antigen bound to the surface of an antigen-presenting cell. The molecular pathways that regulate T cell quiescence remain largely unknown. Here we show that forced expression of the lung Krüppel-like transcription factor (LKLF) in Jurkat T cells is sufficient to program a quiescent phenotype characterized by decreased proliferation, reduced cell size and protein synthesis and decreased surface expression of activation markers. Conversely, LKLF-deficient peripheral T cells produced by gene targeting showed increased proliferation, increased cell size and enhanced expression of surface activation markers in vivo. LKLF appeared to function, at least in part, by decreasing expression of the proto-oncogene encoding c-Myc. Forced expression of LKLF was associated with markedly decreased c-Myc expression. In addition, many effects of LKLF expression were mimicked by expression of the dominant-negative MadMyc protein and rescued by overexpression of c-Myc. Thus, LKLF is both necessary and sufficient to program quiescence in T cells and functions, in part, by negatively regulating a c-Myc--dependent pathway.
Collapse
Affiliation(s)
- A F Buckley
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|