1
|
Zheng C, Zhang C, He Y, Lin S, Zhu Z, Wang H, Chen G. Cbfβ: A key regulator in skeletal stem cell differentiation, bone development, and disease. FASEB J 2025; 39:e70399. [PMID: 39996474 DOI: 10.1096/fj.202500030r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025]
Abstract
The skeletal system comprises closely related yet functionally distinct bone and cartilage tissues, regulated by a complex network of transcriptional factors and signaling molecules. Among these, core-binding factor subunit beta (Cbfβ) emerges as a critical co-transcriptional factor that stabilizes Runx proteins, playing indispensable roles in skeletal development and homeostasis. Emerging evidence from genetic mouse models has highlighted the essential role of Cbfβ in directing the lineage commitment of mesenchymal stem cells (MSCs) and their differentiation into osteoblasts and chondrocytes. Notably, Cbfβ deficiency is strongly associated with severe skeletal dysplasia, affecting both endochondral and intramembranous ossification during embryonic and postnatal development. In this review, we synthesize recent advancements in understanding the structural and molecular functions of Cbfβ, with a particular focus on its interactions with key signaling pathways, including BMP/TGF-β, Wnt/β-catenin, Hippo/YAP, and IHH/PTHrP. These pathways converge on the Cbfβ/RUNX2 complex, which orchestrates a gene expression program essential for osteogenesis, bone formation, and cartilage development. The integration of these signaling networks ensures the precise regulation of skeletal development, remodeling, and repair. Furthermore, the successful local delivery of Cbfβ to address bone abnormalities underscores its potential as a novel therapeutic target for skeletal disorders such as cleidocranial dysplasia, osteoarthritis, and bone metastases. By elucidating the molecular mechanisms underlying Cbfβ function and its interactions with key signaling pathways, these insights not only advance our understanding of skeletal biology but also offer promising avenues for clinical intervention, ultimately improving outcomes for patients with skeletal disorders.
Collapse
Affiliation(s)
- Chenggong Zheng
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Chenyang Zhang
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yiliang He
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Sisi Lin
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhenya Zhu
- Department of Orthopedics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Haidong Wang
- Department of Orthopedics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
2
|
Yoshida T, Yamasaki K, Tadagaki K, Kuwahara Y, Matsumoto A, Sofovic AE, Kondo N, Sakai T, Okuda T. Tumor necrosis factor‑related apoptosis‑inducing ligand is a novel transcriptional target of runt‑related transcription factor 1. Int J Oncol 2021; 60:6. [PMID: 34958111 PMCID: PMC8727134 DOI: 10.3892/ijo.2021.5296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/23/2021] [Indexed: 11/26/2022] Open
Abstract
Runt-related transcription factor 1 (RUNX1), which is also known as acute myeloid leukemia 1 (AML1), has been frequently found with genomic aberrations in human leukemia. RUNX1 encodes a transcription factor that can regulate the expression of hematopoietic genes. In addition, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) performs an important function for malignant tumors in immune surveillance. However, the regulatory mechanism of TRAIL expression remain to be fully elucidated. In the present study, tetradecanoylphorbol 13-acetate-treated megakaryocytic differentiated K562 cells was used to examine the effect of RUNX1 on TRAIL expression. Luciferase assay series of TRAIL promoters for the cells co-transfected with RUNX1 and core-binding factor β (CBFβ) expression vectors were performed to evaluate the nature of TRAIL transcriptional regulation. Electrophoresis mobility shift assay of the RUNX1 consensus sequence of the TRAIL promoter with recombinant RUNX1 and CBFβ proteins was also performed. BloodSpot database analysis for TRAIL expression in patients with acute myeloid leukemia were performed. The expression of TRAIL, its receptor Death receptor 4 and 5 and RUNX1 in K562 cells transfected with the RUNX1 expression vector and RUNX1 siRNA were evaluated by reverse transcription-quantitative PCR (RT-qPCR). TRAIL and RUNX1-ETO expression was also measured in Kasumi-1 cells transfected with RUNX1-ETO siRNA and in KG-1 cells transfected with RUNX1-ETO expression plasmid, both by RT-qPCR. Cell counting, lactate dehydrogenase assay and cell cycle analysis by flow cytometry were performed on Kasumi-1, KG-1, SKNO-1 and K562 cells treated with TRAIL and HDAC inhibitors sodium butyrate or valproic acid. The present study demonstrated that RUNX1 is a transcriptional regulator of TRAIL. It was initially found that the induction of TRAIL expression following the megakaryocytic differentiation of human leukemia cells was RUNX1-dependent. Subsequently, overexpression of RUNX1 was found to increase TRAIL mRNA expression by activating its promoter activity. Additional analyses revealed that RUNX1 regulated the expression of TRAIL in an indirect manner, because RUNX1 retained its ability to activate this promoter following the mutation of all possible RUNX1 consensus sites. Furthermore, TRAIL expression was reduced in leukemia cells carrying the t(8;21) translocation, where the RUNX1-ETO chimeric protein interfere with normal RUNX1 function. Exogenous treatment of recombinant TRAIL proteins was found to induce leukemia cell death. To conclude, the present study provided a novel mechanism, whereby TRAIL is a target gene of RUNX1 and TRAIL expression was inhibited by RUNX1-ETO. These results suggest that TRAIL is a promising agent for the clinical treatment of t(8;21) AML.
Collapse
Affiliation(s)
- Tatsushi Yoshida
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Kenta Yamasaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Kenjiro Tadagaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Yasumichi Kuwahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Akifumi Matsumoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Adèm Ejub Sofovic
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Noriko Kondo
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| | - Tsukasa Okuda
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi‑Hirokoji, Kamigyo‑ku, Kyoto 602‑8566, Japan
| |
Collapse
|
3
|
Robinson SB, Refai O, Hardaway JA, Sturgeon S, Popay T, Bermingham DP, Freeman P, Wright J, Blakely RD. Dopamine-dependent, swimming-induced paralysis arises as a consequence of loss of function mutations in the RUNX transcription factor RNT-1. PLoS One 2019; 14:e0216417. [PMID: 31083672 PMCID: PMC6513266 DOI: 10.1371/journal.pone.0216417] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/21/2019] [Indexed: 11/18/2022] Open
Abstract
Dopamine (DA) is a neurotransmitter with actions across phylogeny that modulate core behaviors such as motor activity, reward, attention, and cognition. Perturbed DA signaling in humans is associated with multiple disorders, including addiction, ADHD, schizophrenia, and Parkinson's disease. The presynaptic DA transporter exerts powerful control on DA signaling by efficient clearance of the neurotransmitter following release. As in vertebrates, Caenorhabditis elegans DAT (DAT-1) constrains DA signaling and loss of function mutations in the dat-1 gene result in slowed crawling on solid media and swimming-induced paralysis (Swip) in water. Previously, we identified a mutant line, vt34, that exhibits robust DA-dependent Swip. vt34 exhibits biochemical and behavioral phenotypes consistent with reduced DAT-1 function though vt34; dat-1 double mutants exhibit an enhanced Swip phenotype, suggesting contributions of the vt34-associated mutation to additional mechanisms that lead to excess DA signaling. SNP mapping and whole genome sequencing of vt34 identified the site of the molecular lesion in the gene B0412.2 that encodes the Runx transcription factor ortholog RNT-1. Unlike dat-1 animals, but similar to other loss of function rnt-1 mutants, vt34 exhibits altered male tail morphology and reduced body size. Deletion mutations in both rnt-1 and the bro-1 gene, which encodes a RNT-1 binding partner also exhibit Swip. Both vt34 and rnt-1 mutations exhibit reduced levels of dat-1 mRNA as well as the tyrosine hydroxylase ortholog cat-2. Although reporter studies indicate that rnt-1 is expressed in DA neurons, its re-expression in DA neurons of vt34 animals fails to fully rescue Swip. Moreover, as shown for vt34, rnt-1 mutation exhibits additivity with dat-1 in generating Swip, as do rnt-1 and bro-1 mutations, and vt34 exhibits altered capacity for acetylcholine signaling at the neuromuscular junction. Together, these findings identify a novel role for rnt-1 in limiting DA neurotransmission and suggest that loss of RNT-1 may disrupt function of both DA neurons and body wall muscle to drive Swip.
Collapse
Affiliation(s)
- Sarah B Robinson
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
| | - J Andrew Hardaway
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Sarah Sturgeon
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Tessa Popay
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Daniel P Bermingham
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Phyllis Freeman
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, United States of America
| | - Jane Wright
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States of America
| |
Collapse
|
4
|
Lie-A-Ling M, Marinopoulou E, Lilly AJ, Challinor M, Patel R, Lancrin C, Kouskoff V, Lacaud G. Regulation of RUNX1 dosage is crucial for efficient blood formation from hemogenic endothelium. Development 2018; 145:dev149419. [PMID: 29530939 PMCID: PMC5868988 DOI: 10.1242/dev.149419] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/30/2018] [Indexed: 12/20/2022]
Abstract
During ontogeny, hematopoietic stem and progenitor cells arise from hemogenic endothelium through an endothelial-to-hematopoietic transition that is strictly dependent on the transcription factor RUNX1. Although it is well established that RUNX1 is essential for the onset of hematopoiesis, little is known about the role of RUNX1 dosage specifically in hemogenic endothelium and during the endothelial-to-hematopoietic transition. Here, we used the mouse embryonic stem cell differentiation system to determine if and how RUNX1 dosage affects hemogenic endothelium differentiation. The use of inducible Runx1 expression combined with alterations in the expression of the RUNX1 co-factor CBFβ allowed us to evaluate a wide range of RUNX1 levels. We demonstrate that low RUNX1 levels are sufficient and necessary to initiate an effective endothelial-to-hematopoietic transition. Subsequently, RUNX1 is also required to complete the endothelial-to-hematopoietic transition and to generate functional hematopoietic precursors. In contrast, elevated levels of RUNX1 are able to drive an accelerated endothelial-to-hematopoietic transition, but the resulting cells are unable to generate mature hematopoietic cells. Together, our results suggest that RUNX1 dosage plays a pivotal role in hemogenic endothelium maturation and the establishment of the hematopoietic system.
Collapse
Affiliation(s)
- Michael Lie-A-Ling
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Elli Marinopoulou
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Andrew J Lilly
- Stem Cell Hematopoiesis, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Mairi Challinor
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Rahima Patel
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Christophe Lancrin
- EMBL Rome, Epigenetics and Neurobiology Unit, Campus Adriano Buzzati-Traverso Via Ramarini 32, 00015 Monterotondo, Italy
| | - Valerie Kouskoff
- Stem Cell Hematopoiesis, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
5
|
Simões B, Conceição N, Matias AC, Bragança J, Kelsh RN, Cancela ML. Molecular characterization of cbfβ gene and identification of new transcription variants: implications for function. Arch Biochem Biophys 2015; 567:1-12. [PMID: 25575784 DOI: 10.1016/j.abb.2014.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 12/09/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
The CBFβ gene encodes a transcription factor that, in combination with CBFα (also called Runx, runt-related transcription factor) regulates expression of several target genes. CBFβ interacts with all Runx family members, such as RUNX2, a regulator of bone-related gene transcription that contains a conserved DNA-binding domain. CBFβ stimulates DNA binding of the Runt domain, and is essential for most of the known functions of RUNX2. A comparative analysis of the zebrafish cbfβ gene and protein, and of its orthologous identified homologous proteins in different species indicates a highly conserved function. We cloned eleven zebrafish cbfβ gene transcripts, one resulting in the known Cbfβ protein (with 187 aa), and three additional variants resulting from skipping exon 5a (resulting in a protein with 174 aa) or exon 5b (resulting in a protein with 201 aa), both observed for the first time in zebrafish, and a completely novel isoform containing both exon 5a and 5b (resulting in a protein with 188 aa). Functional analysis of these isoforms provides insight into their role in regulating gene transcription. From the other variants two are premature termination Cbfβ forms, while the others show in-frame exon-skipping causing changes in the Cbfβ domain that may affect its function.
Collapse
Affiliation(s)
- B Simões
- Department of Biomedical Sciences and Medicine/DCBM, University of Algarve, Faro, Portugal; PhD Program in Biomedical Sciences, University of Algarve, Faro, Portugal; Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - N Conceição
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - A C Matias
- Department of Biomedical Sciences and Medicine/DCBM, University of Algarve, Faro, Portugal; Centre for Molecular and Structural Biomedicine, University of Algarve, Faro, Portugal
| | - J Bragança
- Department of Biomedical Sciences and Medicine/DCBM, University of Algarve, Faro, Portugal; Centre for Molecular and Structural Biomedicine, University of Algarve, Faro, Portugal
| | - R N Kelsh
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Claverton Down, United Kingdom
| | - M L Cancela
- Department of Biomedical Sciences and Medicine/DCBM, University of Algarve, Faro, Portugal; Centre of Marine Sciences, University of Algarve, Faro, Portugal.
| |
Collapse
|
6
|
Nah GSS, Tay BH, Brenner S, Osato M, Venkatesh B. Characterization of the Runx gene family in a jawless vertebrate, the Japanese lamprey (Lethenteron japonicum). PLoS One 2014; 9:e113445. [PMID: 25405766 PMCID: PMC4236176 DOI: 10.1371/journal.pone.0113445] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/24/2014] [Indexed: 01/08/2023] Open
Abstract
The cyclostomes (jawless vertebrates), comprising lampreys and hagfishes, are the sister group of jawed vertebrates (gnathostomes) and are hence an important group for the study of vertebrate evolution. In mammals, three Runx genes, Runx1, Runx2 and Runx3, encode transcription factors that are essential for cell proliferation and differentiation in major developmental pathways such as haematopoiesis, skeletogenesis and neurogenesis and are frequently associated with diseases. We describe here the characterization of Runx gene family members from a cyclostome, the Japanese lamprey (Lethenteron japonicum). The Japanese lamprey contains three Runx genes, RunxA, RunxB, and RunxC. However, phylogenetic and synteny analyses suggest that they are not one-to-one orthologs of gnathostome Runx1, Runx2 and Runx3. The major protein domains and motifs found in gnathostome Runx proteins are highly conserved in the lamprey Runx proteins. Although all gnathostome Runx genes each contain two alternative promoters, P1 (distal) and P2 (proximal), only lamprey RunxB possesses the alternative promoters; lamprey RunxA and RunxC contain only P2 and P1 promoter, respectively. Furthermore, the three lamprey Runx genes give rise to fewer alternative isoforms than the three gnathostome Runx genes. The promoters of the lamprey Runx genes lack the tandem Runx-binding motifs that are highly conserved among the P1 promoters of gnathostome Runx1, Runx2 and Runx3 genes; instead these promoters contain dispersed single Runx-binding motifs. The 3'UTR of lamprey RunxB contains binding sites for miR-27 and miR-130b/301ab, which are conserved in mammalian Runx1 and Runx3, respectively. Overall, the Runx genes in lamprey seem to have experienced a different evolutionary trajectory from that of gnathostome Runx genes which are highly conserved all the way from cartilaginous fishes to mammals.
Collapse
Affiliation(s)
- Giselle Sek Suan Nah
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Boon-Hui Tay
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sydney Brenner
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Motomi Osato
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research, Singapore, Singapore
- * E-mail: (MO); (BV)
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail: (MO); (BV)
| |
Collapse
|
7
|
Nah GSS, Lim ZW, Tay BH, Osato M, Venkatesh B. Runx family genes in a cartilaginous fish, the elephant shark (Callorhinchus milii). PLoS One 2014; 9:e93816. [PMID: 24699678 PMCID: PMC3974841 DOI: 10.1371/journal.pone.0093816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/06/2014] [Indexed: 12/11/2022] Open
Abstract
The Runx family genes encode transcription factors that play key roles in hematopoiesis, skeletogenesis and neurogenesis and are often implicated in diseases. We describe here the cloning and characterization of Runx1, Runx2, Runx3 and Runxb genes in the elephant shark (Callorhinchus milii), a member of Chondrichthyes, the oldest living group of jawed vertebrates. Through the use of alternative promoters and/or alternative splicing, each of the elephant shark Runx genes expresses multiple isoforms similar to their orthologs in human and other bony vertebrates. The expression profiles of elephant shark Runx genes are similar to those of mammalian Runx genes. The syntenic blocks of genes at the elephant shark Runx gene loci are highly conserved in human, but represented by shorter conserved blocks in zebrafish indicating a higher degree of rearrangements in this teleost fish. Analysis of promoter regions revealed conservation of binding sites for transcription factors, including two tandem binding sites for Runx that are totally conserved in the distal promoter regions of elephant shark Runx1-3. Several conserved noncoding elements (CNEs), which are putative cis-regulatory elements, and miRNA binding sites were identified in the elephant shark and human Runx gene loci. Some of these CNEs and miRNA binding sites are absent in teleost fishes such as zebrafish and fugu. In summary, our analysis reveals that the genomic organization and expression profiles of Runx genes were already complex in the common ancestor of jawed vertebrates.
Collapse
Affiliation(s)
- Giselle Sek Suan Nah
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Zhi Wei Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Boon-Hui Tay
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Motomi Osato
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research, Singapore, Singapore
- * E-mail: (MO); (BV)
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail: (MO); (BV)
| |
Collapse
|
8
|
Hultquist JF, McDougle RM, Anderson BD, Harris RS. HIV type 1 viral infectivity factor and the RUNX transcription factors interact with core binding factor β on genetically distinct surfaces. AIDS Res Hum Retroviruses 2012; 28:1543-51. [PMID: 22725134 DOI: 10.1089/aid.2012.0142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) requires the cellular transcription factor core binding factor subunit β (CBFβ) to stabilize its viral infectivity factor (Vif) protein and neutralize the APOBEC3 restriction factors. CBFβ normally heterodimerizes with the RUNX family of transcription factors, enhancing their stability and DNA-binding affinity. To test the hypothesis that Vif may act as a RUNX mimic to bind CBFβ, we generated a series of CBFβ mutants at the RUNX/CBFβ interface and tested their ability to stabilize Vif and impact transcription at a RUNX-dependent promoter. While several CBFβ amino acid substitutions disrupted promoter activity, none of these impacted the ability of CBFβ to stabilize Vif or enhance degradation of APOBEC3G. A mutagenesis screen of CBFβ surface residues identified a single amino acid change, F68D, that disrupted Vif binding and its ability to degrade APOBEC3G. This mutant still bound RUNX and stimulated RUNX-dependent transcription. These separation-of-function mutants demonstrate that HIV-1 Vif and the RUNX transcription factors interact with cellular CBFβ on genetically distinct surfaces.
Collapse
Affiliation(s)
- Judd F. Hultquist
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, Minnesota
| | - Rebecca M. McDougle
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Brett D. Anderson
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Reuben S. Harris
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
9
|
Walrad PB, Hang S, Joseph GS, Salas J, Gergen JP. Distinct contributions of conserved modules to Runt transcription factor activity. Mol Biol Cell 2010; 21:2315-26. [PMID: 20462957 PMCID: PMC2893994 DOI: 10.1091/mbc.e09-11-0953] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
An investigation of the in vivo roles of conserved regions of the Drosophila Runt protein outside of the DNA-binding Runt domain reveals distinct requirements in different regulatory activities. The conserved VWRPY-containing C-terminus required for repression of only a subset of targets is also found to participate in activation of other targets. Runx proteins play vital roles in regulating transcription in numerous developmental pathways throughout the animal kingdom. Two Runx protein hallmarks are the DNA-binding Runt domain and a C-terminal VWRPY motif that mediates interaction with TLE/Gro corepressor proteins. A phylogenetic analysis of Runt, the founding Runx family member, identifies four distinct regions C-terminal to the Runt domain that are conserved in Drosophila and other insects. We used a series of previously described ectopic expression assays to investigate the functions of these different conserved regions in regulating gene expression during embryogenesis and in controlling axonal projections in the developing eye. The results indicate each conserved region is required for a different subset of activities and identify distinct regions that participate in the transcriptional activation and repression of the segmentation gene sloppy-paired-1 (slp1). Interestingly, the C-terminal VWRPY-containing region is not required for repression but instead plays a role in slp1 activation. Genetic experiments indicating that Groucho (Gro) does not participate in slp1 regulation further suggest that Runt's conserved C-terminus interacts with other factors to promote transcriptional activation. These results provide a foundation for further studies on the molecular interactions that contribute to the context-dependent properties of Runx proteins as developmental regulators.
Collapse
Affiliation(s)
- Pegine B Walrad
- Department of Biochemistry and Cell Biology and the Center for Developmental Genetics, Graduate Programs in Molecular and Cellular Biology and Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | | | | | | | | |
Collapse
|
10
|
Lukosz M, Jakob S, Büchner N, Zschauer TC, Altschmied J, Haendeler J. Nuclear redox signaling. Antioxid Redox Signal 2010; 12:713-42. [PMID: 19737086 DOI: 10.1089/ars.2009.2609] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species have been described to modulate proteins within the cell, a process called redox regulation. However, the importance of compartment-specific redox regulation has been neglected for a long time. In the early 1980s and 1990s, many in vitro studies introduced the possibility that nuclear redox signaling exists. However, the functional relevance for that has been greatly disregarded. Recently, it has become evident that nuclear redox signaling is indeed one important signaling mechanism regulating a variety of cellular functions. Transcription factors, and even kinases and phosphatases, have been described to be redox regulated in the nucleus. This review describes several of these proteins in closer detail and explains their functions resulting from nuclear localization and redox regulation. Moreover, the redox state of the nucleus and several important nuclear redox regulators [Thioredoxin-1 (Trx-1), Glutaredoxins (Grxs), Peroxiredoxins (Prxs), and APEX nuclease (multifunctional DNA-repair enzyme) 1 (APEX1)] are introduced more precisely, and their necessity for regulation of transcription factors is emphasized.
Collapse
Affiliation(s)
- Margarete Lukosz
- Molecular Cell & Aging Research, IUF (Institute for Molecular Preventive Medicine), At the University of Duesseldorf gGmbH, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | | | | | | | | | | |
Collapse
|
11
|
Wang CQ, Jacob B, Nah GSS, Osato M. Runx family genes, niche, and stem cell quiescence. Blood Cells Mol Dis 2010; 44:275-86. [PMID: 20144877 DOI: 10.1016/j.bcmd.2010.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 01/05/2010] [Indexed: 02/07/2023]
Abstract
In multicellular organisms, terminally differentiated cells of most tissues are short-lived and therefore require constant replenishment from rapidly dividing stem cells for homeostasis and tissue repair. For the stem cells to last throughout the lifetime of the organism, however, a small subset of stem cells, which are maintained in a hibernation-like state known as stem cell quiescence, is required. Such dormant stem cells reside in the niche and are activated into proliferation only when necessary. A multitude of factors are required for the maintenance of stem cell quiescence and niche. In particular, the Runx family genes have been implicated in stem cell quiescence in various organisms and tissues. In this review, we discuss the maintenance of stem cell quiescence in various tissues, mainly in the context of the Runx family genes, and with special focus on the hematopoietic system.
Collapse
Affiliation(s)
- Chelsia Qiuxia Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
12
|
|
13
|
Sakai E, Kitajima K, Sato A, Nakano T. Increase of hematopoietic progenitor and suppression of endothelial gene expression by Runx1 expression during in vitro ES differentiation. Exp Hematol 2009; 37:334-45. [PMID: 19218012 DOI: 10.1016/j.exphem.2008.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 11/12/2008] [Accepted: 11/13/2008] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Runx1 is essential for both the establishment of hematopoiesis during development and maintenance of adult hematopoiesis. To reveal the roles of Runx1, we examined how and when Runx1 functions during development of hematopoiesis, and revealed the genes controlled by Runx1. MATERIALS AND METHODS A combined in vitro approach involving in vitro hematopoietic differentiation of embryonic stem cells and conditional gene expression of Runx1 was utilized for this study. Then we analyzed the effects of Runx1 on the differentiation and proliferation of hematopoietic cells and carried out DNA microarray analysis. RESULTS Pulse expression of Runx1 prior to the emergence of hematopoietic cells caused immature hematopoietic cell increase but did not have any effects on the induction of hemogenic cells. During this process, the mRNA level of several endothelial cell-specific genes was downregulated. CONCLUSION Runx1 expression play important roles on the proliferation of emerging immature hematopoietic progenitors or the transition process from endothelial to hematopoietic cells presumably by suppressing the genes related to endothelial phenotype.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Pathology, School of Medicine and Frontier Biosciences, Osaka University, Yamadaok, Osaka, Japan.
| | | | | | | |
Collapse
|
14
|
Sullivan JC, Sher D, Eisenstein M, Shigesada K, Reitzel AM, Marlow H, Levanon D, Groner Y, Finnerty JR, Gat U. The evolutionary origin of the Runx/CBFbeta transcription factors--studies of the most basal metazoans. BMC Evol Biol 2008; 8:228. [PMID: 18681949 PMCID: PMC2527000 DOI: 10.1186/1471-2148-8-228] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Accepted: 08/05/2008] [Indexed: 11/17/2022] Open
Abstract
Background Members of the Runx family of transcriptional regulators, which bind DNA as heterodimers with CBFβ, are known to play critical roles in embryonic development in many triploblastic animals such as mammals and insects. They are known to regulate basic developmental processes such as cell fate determination and cellular potency in multiple stem-cell types, including the sensory nerve cell progenitors of ganglia in mammals. Results In this study, we detect and characterize the hitherto unexplored Runx/CBFβ genes of cnidarians and sponges, two basal animal lineages that are well known for their extensive regenerative capacity. Comparative structural modeling indicates that the Runx-CBFβ-DNA complex from most cnidarians and sponges is highly similar to that found in humans, with changes in the residues involved in Runx-CBFβ dimerization in either of the proteins mirrored by compensatory changes in the binding partner. In situ hybridization studies reveal that Nematostella Runx and CBFβ are expressed predominantly in small isolated foci at the base of the ectoderm of the tentacles in adult animals, possibly representing neurons or their progenitors. Conclusion These results reveal that Runx and CBFβ likely functioned together to regulate transcription in the common ancestor of all metazoans, and the structure of the Runx-CBFβ-DNA complex has remained extremely conserved since the human-sponge divergence. The expression data suggest a hypothesis that these genes may have played a role in nerve cell differentiation or maintenance in the common ancestor of cnidarians and bilaterians.
Collapse
Affiliation(s)
- James C Sullivan
- Department of Biology, Boston University, 5 Cummington St, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ng CEL, Osato M, Tay BH, Venkatesh B, Ito Y. cDNA cloning of Runx family genes from the pufferfish (Fugu rubripes). Gene 2007; 399:162-73. [PMID: 17604919 DOI: 10.1016/j.gene.2007.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 05/15/2007] [Accepted: 05/15/2007] [Indexed: 11/18/2022]
Abstract
The Runx family genes are involved in hematopoiesis, osteogenesis and neuropoiesis, and mutations in these genes have been frequently associated with human hereditary diseases and cancers. Here we report the cDNA cloning of the full Runx gene family of the pufferfish (Fugu rubripes), which comprises frRunx1, frRunx2, frRunx3, frRunt and frCbfb. Fugu is evolutionarily distant from mammals, thus the annotation of the frRunx family genes greatly facilitates comparative genomics approaches. Protein sequence comparison revealed that the fugu genes show high conservation in the Runt domain and PY and VWRPY motifs. frRunx1 had an extra stretch of eight histidine residues, while frRunx2 lacked the poly-glutamine/-alanine stretch that is a hallmark of the mammalian Runx2 genes. Analysis of the promoter regions revealed high conservation of the binding sites for transcription factors, including Runx sites in the P1 promoters. Abundant CpG dinucleotides in the P2 promoter regions were also detected. The expression patterns of the frRunx family genes in various tissues showed high similarity to those of the mammalian Runx genes. The genomic structures of the fugu and mammalian Runx genes are largely conserved except for a split exon 2 in frRunx1 and an extra exon in the C-terminal region of frRunx3 that is missing in mammalian Runx3 genes. The similarities and differences between the Runx family genes of fugu and mammals will improve our understanding of the functions of these proteins.
Collapse
Affiliation(s)
- Cherry Ee Lin Ng
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, 138673, Singapore
| | | | | | | | | |
Collapse
|
16
|
Huang C, Ida H, Ito K, Zhang H, Ito Y. Contribution of reactivated RUNX3 to inhibition of gastric cancer cell growth following suberoylanilide hydroxamic acid (vorinostat) treatment. Biochem Pharmacol 2007; 73:990-1000. [PMID: 17276407 DOI: 10.1016/j.bcp.2006.12.013] [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] [Received: 10/23/2006] [Revised: 11/27/2006] [Accepted: 12/11/2006] [Indexed: 01/07/2023]
Abstract
Vorinostat (suberoylanilide hydroxamic acid, SAHA) represents a new class of highly potent histone deacetylase (HDAC) inhibitors that cause growth arrest, differentiation, and apoptosis of many tumor types in vitro and in vivo. RUNX3, a gastric tumor suppressor, is epigenetically silenced in gastric cancer cells. This study investigates the role of RUNX3 in vorinostat-induced suppression of gastric cancer cell growth. RUNX3 was up-regulated by vorinostat in gastric cancer cell lines not expressing RUNX3. In terms of cell viability, the mean IC(50) of vorinostat in RUNX3-negative cells was significantly lower than that seen in RUNX3-positive cells, indicating that the former are more sensitive to vorinostat in terms of growth arrest than are RUNX3-positive lines. The mechanism underlying this difference was found to be reactivation of RUNX3 expression by vorinostat and concomitant increase in acetylated histone H3 in the promoter region of RUNX3. Using three RUNX3-negative cell lines, we determined the contribution of RUNX3 reactivation to growth inhibition and induction of apoptosis following treatment of cells with vorinostat and found that up-regulated RUNX3 was significantly responsible for tumor suppressive activities.
Collapse
Affiliation(s)
- Canhua Huang
- Oncology Research Institute, National University of Singapore, Singapore
| | | | | | | | | |
Collapse
|
17
|
Suad O, Eyal E, Blumenzweig I, Kessler N, Levanon D, Groner Y, Shakked Z. RUN-CBFβ Interaction inC. elegans: Computational Prediction and Experimental Verification. J Biomol Struct Dyn 2007; 24:343-58. [PMID: 17206850 DOI: 10.1080/07391102.2007.10507124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The Runt domain proteins are eukaryotic transcription factors that regulate major developmental pathways. All members of this family contain a highly-conserved sequence-specific DNA binding domain: the Runt domain (RD). Structural and biochemical studies have shown that the Runt domain undergoes a conformational transition upon binding to DNA and that this process is regulated by an unrelated partner protein CBFbeta that enhances the DNA binding affinity of RD. Most of the reported studies on the Runt domain transcription factors were performed on proteins from mammals and Drosophila whereas very little has been known about the C. elegans RD protein, RUN, which provides the simplest model system for understanding the function of this class of transcription factors. We performed computational studies on RD domains from various species including C. elegans, Drosophila, and human, using the atom-atom contact surface area scoring method. The scoring analysis indicates that the DNA binding regulation of the C. elegans RD protein (CeRD) occurs via its interaction with a CBFbeta-like partner, as found for the human proteins, whereas a different mode of regulation may occur in the Drosophila system. Sequence, secondary structure and fold analyses of a putative CBFbeta protein identified in the C. elegans genome, CeCBFbeta, sharing a 22% identity with the human protein, predict a similar structure of this protein to that of the human CBFbeta protein. We produced the C. elegans proteins CeRD and CeCBFbeta in bacteria and confirmed their physical interaction as well as cross interactions with the corresponding human proteins. We also confirmed the structural similarity of CBFbeta and CeCBFbeta by circular dichroism analysis. The combined results suggest that a similar mechanism of regulation operates for the human and the C. elegans RD proteins despite the low sequence identity between their CBFbeta proteins and the evolutionary distance between the two systems.
Collapse
Affiliation(s)
- Oded Suad
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
18
|
Asou N, Yanagida M, Huang L, Yamamoto M, Shigesada K, Mitsuya H, Ito Y, Osato M. Concurrent transcriptional deregulation of AML1/RUNX1 and GATA factors by the AML1-TRPS1 chimeric gene in t(8;21)(q24;q22) acute myeloid leukemia. Blood 2007; 109:4023-7. [PMID: 17244685 DOI: 10.1182/blood-2006-01-031781] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abstract
The Runt domain transcription factor AML1/RUNX1 is essential for the generation of hematopoietic stem cells and is the most frequent target of chromosomal translocations associated with leukemia. Here, we present a new AML1 translocation found in a patient with acute myeloid leukemia M4 with t(8;21)(q24;q22) at the time of relapse. This translocation generated an in-frame chimeric gene consisting of the N-terminal portion of AML1, retaining the Runt domain, fused to the entire length of TRPS1 on the C-terminus. TRPS1 encodes a putative multitype zinc finger (ZF) protein containing 9 C2H2 type ZFs and 1 GATA type ZF. AML1-TRPS1 stimulated proliferation of hematopoietic colony-forming cells and repressed the transcriptional activity of AML1 and GATA-1 by 2 different mechanisms: competition at their cognate DNA-binding sites and physical sequestrations of AML1 and GATA-1, suggesting that simultaneous deregulation of AML1 and GATA factors constitutes a basis for leukemogenesis.
Collapse
MESH Headings
- Animals
- Cell Line
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 21/metabolism
- Chromosomes, Human, Pair 8/genetics
- Chromosomes, Human, Pair 8/metabolism
- Core Binding Factor Alpha 2 Subunit/biosynthesis
- Core Binding Factor Alpha 2 Subunit/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- GATA Transcription Factors/genetics
- GATA Transcription Factors/metabolism
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Oncogene Proteins, Fusion/biosynthesis
- Oncogene Proteins, Fusion/genetics
- Repressor Proteins
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Transcription, Genetic
- Translocation, Genetic/genetics
Collapse
Affiliation(s)
- Norio Asou
- Department of Hematology, Kumamoto University School of Medicine, 1-1-1 Honjo, Kumamoto 860-8556, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Schaubach BM, Wen HY, Kellems RE. Regulation of murine Ada gene expression in the placenta by transcription factor RUNX1. Placenta 2006; 27:269-77. [PMID: 16338472 DOI: 10.1016/j.placenta.2005.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Revised: 03/02/2005] [Accepted: 03/03/2005] [Indexed: 10/25/2022]
Abstract
The formation of the trophoblast cell lineage of the placenta is one of the first developmental events to occur in mammalian embryogenesis. To understand the mechanisms of gene regulation in the trophoblast cell lineage we have used the murine adenosine deaminase gene (Ada) as a model. Ada is highly expressed in trophoblast cells of the placenta and is critical for embryo development. A 770bp fragment of the mouse Ada 5' flanking region is capable of directing trophoblast cell-specific expression in a transgenic model system. Earlier studies identified several critical portions of this fragment, including three footprinting regions that are necessary for correct gene expression in the placenta. Using electromobility shift assays (EMSA), we identified a 5bp sequence within footprint 3 that computer databases predicted bound to the transcription factor RUNX1 (also known as acute myeloid leukemia 1). This prediction was confirmed by supershift analysis using antibodies specific for RUNX1. The functional importance of this binding was demonstrated by both transient transfections and transgenic approaches. A significant reduction in expression of the reporter gene in the placenta was seen when the 5bp RUNX1 binding site was mutated. The findings reported here indicate that the RUNX1 transcription factor plays a significant role in regulating Ada gene expression in the trophoblast cell lineage.
Collapse
Affiliation(s)
- B M Schaubach
- University of Texas at Houston, Biochemistry and Molecular Biology, 6431 Fannin, Houston, TX 77030, USA
| | | | | |
Collapse
|
20
|
Kim HJ, Nam SH, Kim HJ, Park HS, Ryoo HM, Kim SY, Cho TJ, Kim SG, Bae SC, Kim IS, Stein JL, van Wijnen AJ, Stein GS, Lian JB, Choi JY. Four novelRUNX2 mutations including a splice donor site result in the cleidocranial dysplasia phenotype. J Cell Physiol 2006; 207:114-22. [PMID: 16270353 DOI: 10.1002/jcp.20552] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant disorder caused by haploinsufficiency of the RUNX2 gene. In this study, we analyzed by direct sequencing RUNX2 mutations from eleven CCD patients. Four of seven mutations were novel: two nonsense mutations resulted in a translational stop at codon 50 (Q50X) and 112 (E112X); a missense mutation converted arginine to glycine at codon 131 (R131G); and an exon 1 splice donor site mutation (donor splice site GT/AT, IVS1 + 1G > A) at exon 1-intron junction resulted in the deletion of QA stretch contained in exon 1 of RUNX2. We focused on the functional analysis of the IVS1 + 1G > A mutation. A full-length cDNA of this mutation was cloned (RUNX2Deltae1) and expressed in Chinese hamster ovary (CHO) and HeLa cells. Functional analysis of RUNX2Deltae1 was performed with respect to protein stability, nuclear localization, DNA binding, and transactivation activity of a downstream RUNX2 target gene. Protein stability of RUNX2Deltae1 is similar to wild-type RUNX2 as determined by Western blot analysis. Subcellular localization of RUNX2Deltae1, assessed by in situ immunofluorescent staining, was observed with partial retention in both the nucleus and cytoplasm. This finding is in contrast to RUNX2 wild-type, which is detected exclusively in the nucleus. DNA binding activity was also compromised by the RUNX2Deltae1 in gel shift assay. Finally, RUNX2Deltae1 blocked transactivation of the osteocalcin gene determined by transient transfection assay. Our findings demonstrate for the first time that the CCD phenotype can be caused by a splice site mutation, which results in the deletion of N-terminus amino acids containing the QA stretch in RUNX2 that contains a previously unidentified second nuclear localization signal (NLS). We postulate that the QA sequence unique to RUNX2 contributes to a competent structure of RUNX2 that is required for nuclear localization, DNA binding, and transactivation function.
Collapse
Affiliation(s)
- Hyo-Jin Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kagoshima H, Sawa H, Mitani S, Bürglin TR, Shigesada K, Kohara Y. The C. elegans RUNX transcription factor RNT-1/MAB-2 is required for asymmetrical cell division of the T blast cell. Dev Biol 2005; 287:262-73. [PMID: 16226243 DOI: 10.1016/j.ydbio.2005.08.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 08/17/2005] [Accepted: 08/18/2005] [Indexed: 10/25/2022]
Abstract
The RUNX genes encode conserved transcription factors, which play vital roles in the development of various animals and human diseases. Drosophila runt is a secondary pair-rule gene, which regulates embryo segmentation. Human RUNX1, previously known as AML1, is essential for hematopoiesis. C. elegans rnt-1 is co-orthologous to the human RUNX genes. We found that RNT-1Colon, two colonsGFP is expressed in the H0-2, V1-6, and T blast cells in the embryo, and predominantly in the seam cells during larval to adult stages. rnt-1 mutants exhibit a loss of polarity in the asymmetrical T cell division in hermaphrodites and abnormal ray morphology in the male tail. Genetic and molecular analysis revealed that rnt-1 is allelic to mab-2. Mutant analysis suggested that rnt-1/mab-2 is involved in regulating T blast cell polarity in cooperation with the Wnt signaling pathway. Expression studies of GFPColon, two colonsPOP-1 and TLP-1Colon, two colonsGFP reporters in rnt-1/mab-2 mutants indicated that this gene functions upstream of tlp-1 and downstream, or in parallel to, pop-1 in the genetic cascade that controls asymmetry of the T cell division. All our data suggest that RNT-1/MAB-2 functions with POP-1 to control the asymmetry of the T cell division.
Collapse
Affiliation(s)
- Hiroshi Kagoshima
- Genome Biology Laboratory, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan.
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
Runt-related (RUNX) gene family is composed of three members, RUNX1/AML1, RUNX2 and RUNX3, and encodes the DNA-binding (alpha) subunits of the Runt domain transcription factor polyomavirus enhancer-binding protein 2 (PEBP2)/core-binding factor (CBF), which is a heterodimeric transcription factor. RUNX1 is most frequently involved in human acute leukemia. RUNX2 shows oncogenic potential in mouse experimental system. RUNX3 is a strong candidate as a gastric cancer tumor suppressor. The beta subunit gene of PEBP2/CBF is also frequently involved in chromosome rearrangements associated with human leukemia. In this Overview, I will summarize how this growing field has been formed and what are the challenging new frontiers for better understanding of the oncogenic potential of this gene family.
Collapse
Affiliation(s)
- Yoshiaki Ito
- Institute of Molecular and Cell Biology and Oncology Research Institute, National University of Singapore, 30 Medical Drive, Singapore 117609, Singapore.
| |
Collapse
|
23
|
Nakao M, Horiike S, Fukushima-Nakase Y, Nishimura M, Fujita Y, Taniwaki M, Okuda T. Novel loss-of-function mutations of the haematopoiesis-related transcription factor, acute myeloid leukaemia 1/runt-related transcription factor 1, detected in acute myeloblastic leukaemia and myelodysplastic syndrome. Br J Haematol 2004; 125:709-19. [PMID: 15180860 DOI: 10.1111/j.1365-2141.2004.04966.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AML1/RUNX1, which encodes a transcription factor essential for definitive haematopoiesis, is a frequent target of leukaemia-associated chromosome translocations. Point mutations of this gene have also recently been associated with leukaemia and myelodysplastic syndrome (MDS). To further define the frequency and biological characteristics of AML1 mutations, we have examined 170 cases of such diseases. Mutations within the runt-domain were identified in five cases: one of de novo acute myeloid leukaemia (AML) and four of MDS. Where multiple time point samples were available, mutations were detected in the earliest samples, which persisted throughout the disease course. Of the five mutations, one was a silent mutation, two were apparent loss-of-function mutations caused by N-terminal truncation, and two were insertions, I150ins and K168ins, which preserved most of the AML1 DNA-binding domain. Both AML1 molecules with insertion mutations were non-functional in that they were unable to rescue haematological defects in AML1-deficient mouse embryonic stem cells. In addition, activating mutations of N-ras, deletion of chromosome 12p, or inactivation of TP53 accompanied some of the AML1 mutations. Together, these observations strongly suggest that one-allele inactivation of AML1 serves as an initial or early event that plays an important role in the eventual development of overt diseases with additional genetic alterations.
Collapse
Affiliation(s)
- Mitsushige Nakao
- Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramach-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Huang G, Shigesada K, Wee HJ, Liu PP, Osato M, Ito Y. Molecular basis for a dominant inactivation of RUNX1/AML1 by the leukemogenic inversion 16 chimera. Blood 2004; 103:3200-7. [PMID: 15070703 DOI: 10.1182/blood-2003-07-2188] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe Runt domain transcription factor, PEBP2/CBF, is a heterodimer composed of 2 subunits. The DNA-binding α subunit, or RUNX protein, interacts with a partner PEBP2β/CBFβ through the evolutionarily conserved Runt domain. Each of the genes encoding RUNX1 and PEBP2β/CBFβ is frequently involved in acute myeloid leukemia. The chimeric protein, CBFβ(PEBP2β)/SMMHC, is generated as a result of inversion of chromosome 16 in such a way to retain the heterodimerization domain of PEBP2β at the amino-terminal side fused to the C-terminal coiled-coil region of smooth muscle myosin heavy chain (SMMHC). Here we show that, in the chimeric protein, the second heterodimerization domain is created by the fusion junction, enabling the chimeric protein to interact with RUNX1 at far greater affinity than PEBP2β and inactivate the RUNX1/AML1 function. To explain why and how heterozygous CBFB/MYH11 can inactivate homozygous RUNX1 near to completion, we propose a new model for this chimeric protein that consists of a Y-shaped dimer with unpaired N-terminal halves followed by a coiled-coil for the C-terminal region. (Blood. 2004;103:3200-3207)
Collapse
MESH Headings
- Animals
- Binding Sites/genetics
- Cell Line, Tumor
- Chromosome Inversion
- Core Binding Factor Alpha 2 Subunit
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dimerization
- Genes, Dominant
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Oncogene Proteins, Fusion/chemistry
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Protein Structure, Tertiary
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/chemistry
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Recombinant Fusion Proteins/antagonists & inhibitors
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Sequence Deletion
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/chemistry
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Gang Huang
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Di Leva F, Ferrante MI, Demarchi F, Caravelli A, Matarazzo MR, Giacca M, D'Urso M, D'Esposito M, Franzé A. Human synaptobrevin-like 1 gene basal transcription is regulated through the interaction of selenocysteine tRNA gene transcription activating factor-zinc finger 143 factors with evolutionary conserved cis-elements. J Biol Chem 2003; 279:7734-9. [PMID: 14672948 DOI: 10.1074/jbc.m308140200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The synaptobrevin-like 1 (SYBL1) gene is ubiquitously expressed and codes for an unusual member of the v-SNAREs molecules implicated in cellular exocytosis. This X-linked gene has the peculiarity of also being present on the Y chromosome in a transcriptional inactive status. Moreover, although ubiquitous, the function of SYBL1 is prominent in specific tissues, such as brain. As a first insight into the molecular mechanisms controlling SYBL1 expression, in this report we describe the extent and role of SYBL1 upstream regions and characterize the binding of trans-acting factors. In vivo foot-printing experiments identify three protected regions. Band shift and transient reporter gene assays indicate a strong role of two of these evolutionary conserved regions in regulating SYBL1 transcription. Because one site is the classical CAAT box, we characterized the binding to the other site of the mammalian homologues of the selenocysteine tRNA gene transcription activating factor (Staf) family, zinc-finger transcription factors, and their role in regulating SYBL1 expression. The results reported here clarify that a Staf-zinc finger family factor, together with the CAAT factor, is the major nuclear protein bound to the SYBL1 promoter region and is responsible for its regulation in HeLa cells, thus identifying the basic control of SYBL1 transcription. In vivo binding of Staf proteins to the SYBL1 promoter is confirmed by chromatin immunoprecipitation assays. Our results identify a fourth mRNA promoter stimulated by a member of the Staf-zinc finger family, the function of which on mRNA polymerase II promoters is still very poorly understood.
Collapse
Affiliation(s)
- Francesca Di Leva
- Institute of Genetics and Biophysics A. Buzzati Traverso, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Denkinger DJ, Kawahara RS. Mutex: a method for simultaneous footprinting and determination of base pair specificity for transcription factor binding sites. Anal Biochem 2003; 321:142-5. [PMID: 12963067 DOI: 10.1016/s0003-2697(03)00404-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Diane J Denkinger
- Department of Pharmacology, University of Nebraska Medical Center, 986260 Nebraska Medical Center, Omaha, NE 68198-6260, USA
| | | |
Collapse
|
27
|
Kim S, Koga T, Isobe M, Kern BE, Yokochi T, Chin YE, Karsenty G, Taniguchi T, Takayanagi H. Stat1 functions as a cytoplasmic attenuator of Runx2 in the transcriptional program of osteoblast differentiation. Genes Dev 2003; 17:1979-91. [PMID: 12923053 PMCID: PMC196253 DOI: 10.1101/gad.1119303] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2003] [Accepted: 06/24/2003] [Indexed: 12/21/2022]
Abstract
Bone remodeling is central to maintaining the integrity of the skeletal system, wherein the developed bone is constantly renewed by the balanced action of osteoblastic bone formation and osteoclastic bone resorption. In the present study, we demonstrate a novel function of the Stat1 transcription factor in the regulation of bone remodeling. In the bone of the Stat1-deficient mice, excessive osteoclastogenesis is observed, presumably caused by a loss of negative regulation of osteoclast differentiation by interferon (IFN)-beta. However, the bone mass is unexpectedly increased in these mice. This increase is caused by excessive osteoblast differentiation, wherein Stat1 function is independent of IFN signaling. Actually, Stat1 interacts with Runx2 in its latent form in the cytoplasm, thereby inhibiting the nuclear localization of Runx2, an essential transcription factor for osteoblast differentiation. The new function of Stat1 does not require the Tyr 701 that is phosphorylated when Stat1 becomes a transcriptional activator. Our study provides a unique example in which a latent transcription factor attenuates the activity of another transcription factor in the cytoplasm, and reveals a new regulatory mechanism in bone remodeling.
Collapse
Affiliation(s)
- Sunhwa Kim
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bristow CAP, Shore P. Transcriptional regulation of the human MIP-1alpha promoter by RUNX1 and MOZ. Nucleic Acids Res 2003; 31:2735-44. [PMID: 12771199 PMCID: PMC156734 DOI: 10.1093/nar/gkg401] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The transcription factor RUNX1 (AML-1, PEBP2alphaB and CBFA2) is essential for definitive haematopoiesis, and chromosomal translocations involving the RUNX1 gene are frequently found in acute leukaemias. The gene encoding the histone acetyltransferase MOZ is also rearranged in some acute leukaemias, resulting in the expression of MOZ fusion proteins. MOZ has recently been shown to interact directly with RUNX1, indicating that MOZ fusion proteins act by deregulating RUNX1 function. Macrophage inflammatory protein-1alpha (MIP-1alpha) is a proinflammatory cytokine that also inhibits proliferation of haematopoietic stem cells. Amongst the conserved sequence elements in the human MIP-1alpha promoter are two consensus RUNX sites. We have investigated the role of these RUNX sites in the regulation of the MIP-1alpha promoter by PMA/PHA stimulation in Jurkat T-cells. RUNX1 can specifically bind to both RUNX sites in vitro and chromatin immunoprecipitation assays demonstrated that endogenous RUNX1 is constitutively bound to the endogenous MIP-1alpha promoter. Mutation of the RUNX sites demonstrated that the proximal RUNX site is essential for PMA/PHA-stimulated activation of the MIP-1alpha promoter. Activation of the promoter can also be inhibited by heterologous expression of the repressor protein AML-1/ETO. We further demonstrate that MOZ can activate the MIP-1alpha promoter and that this activation is largely dependent upon the proximal RUNX site. Moreover, we show that co-expression of MOZ and RUNX1 can synergistically activate the MIP-1alpha promoter. The regulation of MIP-1alpha expression by RUNX1/MOZ is discussed in the context of MIP-1alpha's role as an inhibitor of haematopoietic stem cell proliferation and its potential importance in leukaemias associated with RUNX1 or MOZ chromosomal rearrangements.
Collapse
Affiliation(s)
- Claire A P Bristow
- School of Biological Sciences, University of Manchester, 2.205, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | | |
Collapse
|
29
|
Yoshida T, Kanegane H, Osato M, Yanagida M, Miyawaki T, Ito Y, Shigesada K. Functional analysis of RUNX2 mutations in cleidocranial dysplasia: novel insights into genotype-phenotype correlations. Blood Cells Mol Dis 2003; 30:184-93. [PMID: 12732182 DOI: 10.1016/s1079-9796(03)00020-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cleidocranial dysplasia (CCD) is an inherited autosomal-dominant skeletal disease caused by heterozygous mutations in the osteoblast-specific transcription factor, RUNX2. We have performed mutational analysis of RUNX2 on 24 unrelated patients with CCD. In 17 patients, 16 distinct mutations were detected in the coding region of RUNX2: 4 frameshift, 3 nonsense, 6 missense, and 2 splicing mutations alongside one polymorphism. The missense mutations were all clustered within the Runt domain and their protein products showed neither DNA binding nor transactivation. On the other hand, some mutant RUNX2 had the Runt domain intact and remained partially competent for transactivation. Coincidentally, one important phenotype of CCD, the short stature, was significantly milder in the patients with the intact Runt domain than those without. Furthermore, a remarkable correlation was found between the short stature and the number of supernumerary teeth. On the other hand, the classic CCD phenotype, hypoplastic clavicles or open fontanelles, was invariably observed regardless of the degree of short stature or supernumerary teeth. Overall, these results suggest that CCD could result from a much smaller loss in the RUNX2 function than envisioned on the basis of the conventional haploinsufficiency model. This makes an interesting contrast to the case of familial and sporadic leukemias mediated by RUNX1 mutations, in which mutants acting in a dominant negative manner have been suggested to confer a higher propensity to develop leukemia.
Collapse
Affiliation(s)
- Taketoshi Yoshida
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama 930-0194, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Asou N. The role of a Runt domain transcription factor AML1/RUNX1 in leukemogenesis and its clinical implications. Crit Rev Oncol Hematol 2003; 45:129-50. [PMID: 12604126 DOI: 10.1016/s1040-8428(02)00003-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A Runt domain transcription factor AML1/RUNX1 is essential for generation and differentiation of definitive hematopoietic stem cells. AML1 is the most frequent target of chromosomal translocations in acute leukemias. Several chimeric proteins such as AML1-MTG8 and TEL-AML1 have transdominant properties for wild-type AML1 and acts as transcriptional repressors. The transcriptional repression in AML1 fusion proteins is mediated by recruitment of nuclear corepressor complex that maintains local histone deacetylation. Inhibition of the expression of AML1-responsive genes leads to a block in hematopoietic cell differentiation and consequent leukemic transformation. On the other hand, mutations in the Runt domain of the AML1 are identified in both sporadic acute myeloblastic leukemia (AML) without AML1 translocation and familial platelet disorder with predisposition to AML. These observations indicate that a decrease in AML1 dosage resulting from chromosomal translocations or mutations contributes to leukemogenesis. Furthermore, dysregulated chromatin remodeling and transcriptional control appears to be a common pathway in AML1-associated leukemias that could be an important target for the development of new therapeutic agents.
Collapse
Affiliation(s)
- Norio Asou
- Department of Internal Medicine II, Kumamoto University School of Medicine, 1-1-1 Honjo, 860-8556, Kumamoto, Japan.
| |
Collapse
|
31
|
Silva FPG, Morolli B, Storlazzi CT, Anelli L, Wessels H, Bezrookove V, Kluin-Nelemans HC, Giphart-Gassler M. Identification of RUNX1/AML1 as a classical tumor suppressor gene. Oncogene 2003; 22:538-47. [PMID: 12555067 DOI: 10.1038/sj.onc.1206141] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Based on our previous results indicating the presence of a tumor suppressor gene (TSG), chromosome 21 was analysed for loss of heterozygosity (LOH) in 18 patients with acute myeloid leukemia (17, AML-M0; one, AML-M1). Allelotyping at polymorphic loci was performed on purified material, allowing unequivocal detection of allelic loss and homozygous deletions. Six AML-M0 patients shared a common region of LOH harboring a single gene: RUNX1 (AML1), the most frequent site of translocations in acute leukemia and a well-known fusion oncogene. Fluorescence in situ hybridization allowed the identification of deletions with breakpoints within RUNX1 in two patients as the cause of LOH. In the four others the LOH pattern and the presence of two karyotypically normal chromosomes 21 were in line with mitotic recombination. Further molecular and cytogenetic analyses showed that this caused homozygosity of primary RUNX1 mutations: two point mutations, a partial deletion and, most significantly, a complete deletion of RUNX1. These findings identify RUNX1 as a classical TSG: both alleles are mutated or absent in cancer cells from four of the 17 AML-M0 patients examined. In contrast to AML-M0, the AML-M1 patient was trisomic for chromosome 21 and has two mutated and one normal RUNX1 allele, suggesting that the order of mutagenic events leading to leukemia may influence the predominant tumor type.
Collapse
Affiliation(s)
- Fernando P G Silva
- Department of Radiation Genetics and Chemical Mutagenesis, Leiden University Medical Center, P.O. Box 9503, 2300RA Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kundu M, Javed A, Jeon JP, Horner A, Shum L, Eckhaus M, Muenke M, Lian JB, Yang Y, Nuckolls GH, Stein GS, Liu PP. Cbfbeta interacts with Runx2 and has a critical role in bone development. Nat Genet 2002; 32:639-44. [PMID: 12434156 DOI: 10.1038/ng1050] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2002] [Accepted: 10/25/2002] [Indexed: 12/27/2022]
Abstract
Runx2 (runt-related transcription factor 2, also known as Cbfa1, Osf2 and AML3) is essential for bone development in mice, and mutations in RUNX2 are found in 65-80% of individuals with cleidocranial dysplasia. Although all Runx family members can interact with Cbfbeta (core-binding factor b, encoded by Cbfb), a role for Cbfbeta in bone development has not been demonstrated owing to lethality in Cbfb(-/-) mouse embryos at 12.5 days post coitum (d.p.c.) from hemorrhages and lack of definitive hematopoiesis. Using a 'knock-in' strategy, we generated mouse embryonic stem (ES) cells that express Cbfb fused in-frame to a cDNA encoding green fluorescent protein (GFP). Cbfb(+/GFP) mice had normal life spans and appeared normal, but Cbfb(GFP/GFP) pups died within the first day after birth. The Cbfb(GFP/GFP) mice exhibited a delay in endochondral and intramembranous ossification as well as in chondrocyte differentiation, similar to but less severe than delays observed in Runx2(-/-) mice. We demonstrate that Cbfbeta is expressed in developing bone and forms a functional interaction with Runx2, and that Cbfb(GFP) is a hypomorphic allele. The fusion allele maintains sufficient function in hematopoietic cells to bypass the early embryonic lethality, and identifies a new role for Cbfb in bone development. Our findings raise the possibility that mutations in CBFB may be responsible for some cases of cleidocranial dysplasia that are not linked to mutations in RUNX2.
Collapse
Affiliation(s)
- Mondira Kundu
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 49 Convent Drive, Building 49, Room 3A26, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Burns CE, DeBlasio T, Zhou Y, Zhang J, Zon L, Nimer SD. Isolation and characterization of runxa and runxb, zebrafish members of the runt family of transcriptional regulators. Exp Hematol 2002; 30:1381-9. [PMID: 12482499 DOI: 10.1016/s0301-472x(02)00955-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The AML/RUNX family of transcription factors plays important roles in hematopoiesis, neurogenesis, bone development, and segmentation in vertebrate embryos. The aim of this study was to isolate runt-related genes in a genetically and embryologically exploitable system, the zebrafish, and characterize their function during hematopoietic development. MATERIALS AND METHODS Two runt-related genes were isolated by degenerate PCR and standard library screening, and a radiation hybrid panel, T51 RH, was used to resolve their chromosomal localization. In situ hybridization demonstrated their expression whereas their transcriptional activity was assessed using an AML1-responsive reporter gene in the MLA 144 T-cell line. RESULTS We isolated the zebrafish runxa and runxb cDNAs, which encode proteins highly homologous to the human and murine Runx1 (AML1) and Runx3 (AML2) proteins. In contrast to a recent report, we detected runxa expression in both hematopoietic and neural tissues of the developing zebrafish. runxa transcripts first appear during segmentation in bilateral mesodermal cells that coexpress one of the earliest blood and endothelial cell markers, scl/tal-1. By 24 hours postfertilization (hpf), runxa transcripts are seen in the ventral wall of the dorsal aorta. Hematopoietic runxa expression is lost in cloche mutants, which are defective in blood and endothelial cell formation. runxb transcripts are seen in nonhematopoietic domains. Both Runxa and Runxb transactivate an AML1-responsive human promoter in hematopoietic cells. Genomic localization studies demonstrate that runxa is located on linkage group 1 (LG1), and the runxb gene is located on LG13. CONCLUSIONS Our gene expression analysis strongly suggests that both the functional and spatial aorta-gonad-mesonephros (AGM) region has been conserved throughout evolution. Our runxa spatiotemporal expression data shed light on the role of vertebrate Runx1/AML1 in primitive vs definitive hematopoietic development.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Aorta/embryology
- Core Binding Factor Alpha 2 Subunit
- Core Binding Factor Alpha 3 Subunit
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- DNA-Binding Proteins/genetics
- Embryo, Nonmammalian
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/embryology
- Gene Expression Regulation, Developmental
- Genetic Linkage
- Gonads/embryology
- Hematopoiesis/genetics
- Humans
- Mesonephros/embryology
- Molecular Sequence Data
- Phylogeny
- Promoter Regions, Genetic/drug effects
- Proto-Oncogene Proteins
- RNA, Messenger/analysis
- Sequence Homology, Amino Acid
- Transcription Factors/genetics
- Transcription Factors/isolation & purification
- Transcription Factors/pharmacology
- Zebrafish
- Zebrafish Proteins/genetics
- Zebrafish Proteins/isolation & purification
Collapse
|
34
|
Kummalue T, Lou J, Friedman AD. Multimerization via its myosin domain facilitates nuclear localization and inhibition of core binding factor (CBF) activities by the CBFbeta-smooth muscle myosin heavy chain myeloid leukemia oncoprotein. Mol Cell Biol 2002; 22:8278-91. [PMID: 12417730 PMCID: PMC134059 DOI: 10.1128/mcb.22.23.8278-8291.2002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In CBFbeta-SMMHC, core binding factor beta (CBFbeta) is fused to the alpha-helical rod domain of smooth muscle myosin heavy chain (SMMHC). We generated Ba/F3 hematopoietic cells expressing a CBFbeta-SMMHC variant lacking 28 amino acids homologous to the assembly competence domain (ACD) required for multimerization of skeletal muscle myosin. CBFbeta-SMMHC(DeltaACD) multimerized less effectively than either wild-type protein or a variant lacking a different 28-residue segment. In contrast to the control proteins, the DeltaACD mutant did not inhibit CBF DNA binding, AML1-mediated reporter activation, or G(1) to S cell cycle progression, the last being dependent upon activation of CBF-regulated genes. We also linked the CBFbeta domain to 149 or 83 C-terminal CBFbeta-SMMHC residues, retaining 86 or 20 amino acids N-terminal to the ACD. CBFbeta-SMMHC(149C) multimerized and slowed Ba/F3 proliferation, whereas CBFbeta-SMMHC(83C) did not. The majority of CBFbeta-SMMHC and CBFbeta-SMMHC(149C) was detected in the nucleus, whereas the DeltaACD and 83C variants were predominantly cytoplasmic, indicating that multimerization facilitates nuclear retention of CBFbeta-SMMHC. When linked to the simian virus 40 nuclear localization signal (NLS), a significant fraction of CBFbeta-SMMHC(DeltaACD) entered the nucleus but only mildly inhibited CBF activities. As NLS-CBFbeta-SMMHC(83C) remained cytoplasmic, we directed the ACD to CBF target genes by linking it to the AML1 DNA binding domain or to full-length AML1. These AML1-ACD fusion proteins did not affect Ba/F3 proliferation, in contrast to AML1-ETO, which markedly slowed G(1) to S progression dependent upon the integrity of its DNA-binding domain. Thus, the ACD facilitates inhibition of CBF by mediating multimerization of CBFbeta-SMMHC in the nucleus. Therapeutics targeting the ACD may be effective in acute myeloid leukemia cases associated with CBFbeta-SMMHC expression.
Collapse
Affiliation(s)
- Tanawan Kummalue
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | | | | |
Collapse
|
35
|
Yoshida T, Kanegane H, Osato M, Yanagida M, Miyawaki T, Ito Y, Shigesada K. Functional analysis of RUNX2 mutations in Japanese patients with cleidocranial dysplasia demonstrates novel genotype-phenotype correlations. Am J Hum Genet 2002; 71:724-38. [PMID: 12196916 PMCID: PMC378531 DOI: 10.1086/342717] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2002] [Accepted: 06/25/2002] [Indexed: 12/18/2022] Open
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant heritable skeletal disease caused by heterozygous mutations in the osteoblast-specific transcription factor RUNX2. We have performed mutational analysis of RUNX2 on 24 unrelated patients with CCD. In 17 patients, 16 distinct mutations were detected in the coding region of RUNX2: 4 frameshift, 3 nonsense, 6 missense, and 2 splicing mutations, in addition to 1 polymorphism. The missense mutations were all clustered within the Runt domain, and their protein products were severely impaired in DNA binding and transactivation. In contrast, two RUNX2 mutants had the Runt domain intact and remained partially competent for transactivation. One criterion of CCD, short stature, was much milder in the patients with the intact Runt domain than in those without. Furthermore, a significant correlation was found between short stature and the number of supernumerary teeth. On the one hand, these genotype-phenotype correlations highlight a general, quantitative dependency, by skeleto-dental developments, on the gene dosage of RUNX2, which has hitherto been obscured by extreme clinical diversities of CCD; this gene-dosage effect is presumed to manifest on small reductions in the total RUNX2 activity, by approximately one-fourth of the normal level at minimum. On the other hand, the classic CCD phenotype, hypoplastic clavicles or open fontanelles, was invariably observed in all patients, including those with normal height. Thus, the cleidocranial bone formation, as mediated by intramembranous ossification, may require a higher level of RUNX2 than does skeletogenesis (mediated by endochondral ossification), as well as odontogenesis (involving still different complex processes). Overall, these results suggest that CCD could result from much smaller losses in the RUNX2 function than has been envisioned on the basis of the conventional haploinsufficiency model.
Collapse
Affiliation(s)
- Taketoshi Yoshida
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Hirokazu Kanegane
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Motomi Osato
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Masatoshi Yanagida
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Toshio Miyawaki
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Yoshiaki Ito
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| | - Katsuya Shigesada
- Department of Pediatrics, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan; and Departments of Genetics and Molecular Biology and Viral Oncology, Institute for Virus Research, Kyoto University, Kyoto
| |
Collapse
|
36
|
Bäckström S, Wolf-Watz M, Grundström C, Härd T, Grundström T, Sauer UH. The RUNX1 Runt domain at 1.25A resolution: a structural switch and specifically bound chloride ions modulate DNA binding. J Mol Biol 2002; 322:259-72. [PMID: 12217689 DOI: 10.1016/s0022-2836(02)00702-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The evolutionarily conserved Runt homology domain is characteristic of the RUNX family of heterodimeric eukaryotic transcription factors, including RUNX1, RUNX2 and RUNX3. The genes for RUNX1, also termed acute myeloid leukemia protein 1, AML1, and its dimerization partner core-binding factor beta, CBFbeta, are essential for hematopoietic development and are together the most common targets for gene rearrangements in acute human leukemias. Here, we describe the crystal structure of the uncomplexed RUNX1 Runt domain at 1.25A resolution and compare its conformation to previously published structures in complex with DNA, CBFbeta or both. We find that complex formation induces significant structural rearrangements in this immunoglobulin (Ig)-like DNA-binding domain. Most pronounced is the movement of loop L11, which changes from a closed conformation in the free Runt structure to an open conformation in the CBFbeta-bound and DNA-bound forms. This transition, which we refer to as the S-switch, and accompanying structural movements that affect other parts of the Runt domain are crucial for sustained DNA binding. The closed to open transition can be induced by CBFbeta alone; suggesting that one role of CBFbeta is to trigger the S-switch and to stabilize the Runt domain in a conformation enhanced for DNA binding.A feature of the Runt domain hitherto unobserved in any Ig-like DNA-binding domain is the presence of two specifically bound chloride ions. One chloride ion is coordinated by amino acid residues that make direct DNA contact. In a series of electrophoretic mobility-shift analyses, we demonstrate a chloride ion concentration-dependent stimulation of the DNA-binding activity of Runt in the physiological range. A comparable DNA-binding stimulation was observed for negatively charged amino acid residues. This suggests a regulatory mechanism of RUNX proteins through acidic amino acid residues provided by activation domains during cooperative interaction with other transcription factors.
Collapse
Affiliation(s)
- Stefan Bäckström
- Biocrystallography Group, Umeå Centre for Molecular Pathogenesis (UCMP), Umeå University, SE-901 87 Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
37
|
Swarthout JT, D'Alonzo RC, Selvamurugan N, Partridge NC. Parathyroid hormone-dependent signaling pathways regulating genes in bone cells. Gene 2002; 282:1-17. [PMID: 11814673 DOI: 10.1016/s0378-1119(01)00798-3] [Citation(s) in RCA: 241] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Parathyroid hormone (PTH) is an 84-amino-acid polypeptide hormone functioning as a major mediator of bone remodeling and as an essential regulator of calcium homeostasis. PTH and PTH-related protein (PTHrP) indirectly activate osteoclasts resulting in increased bone resorption. During this process, PTH changes the phenotype of the osteoblast from a cell involved in bone formation to one directing bone resorption. In addition to these catabolic effects, PTH has been demonstrated to be an anabolic factor in skeletal tissue and in vitro. As a result, PTH has potential medical application to the treatment of osteoporosis, since intermittent administration of PTH stimulates bone formation. Activation of osteoblasts by PTH results in expression of genes important for the degradation of the extracellular matrix, production of growth factors, and stimulation and recruitment of osteoclasts. The ability of PTH to drive changes in gene expression is dependent upon activation of transcription factors such as the activator protein-1 family, RUNX2, and cAMP response element binding protein (CREB). Much of the regulation of these processes by PTH is protein kinase A (PKA)-dependent. However, while PKA is linked to many of the changes in gene expression directed by PTH, PKA activation has been shown to inhibit mitogen-activated protein kinase (MAPK) and proliferation of osteoblasts. It is now known that stimulation of MAPK and proliferation by PTH at low concentrations is protein kinase C (PKC)-dependent in both osteoblastic and kidney cells. Furthermore, PTH has been demonstrated to regulate components of the cell cycle. However, whether this regulation requires PKC and/or extracellular signal-regulated kinases or whether PTH is able to stimulate other components of the cell cycle is unknown. It is possible that stimulation of this signaling pathway by PTH mediates a unique pattern of gene expression resulting in proliferation in osteoblastic and kidney cells; however, specific examples of this are still unknown. This review will focus on what is known about PTH-mediated cell signaling, and discuss the established or putative PTH-regulated pattern of gene expression in osteoblastic cells following treatment with catabolic (high) or anabolic (low) concentrations of the hormone.
Collapse
Affiliation(s)
- John T Swarthout
- Cell and Molecular Biology Program, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | | | | | | |
Collapse
|
38
|
Nasiadka A, Dietrich BH, Krause HM. Anterior-posterior patterning in the Drosophila embryo. GENE EXPRESSION AT THE BEGINNING OF ANIMAL DEVELOPMENT 2002. [DOI: 10.1016/s1569-1799(02)12027-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
39
|
Wolf-Watz M, Grundström T, Härd T. Structure and backbone dynamics of Apo-CBFbeta in solution. Biochemistry 2001; 40:11423-32. [PMID: 11560490 DOI: 10.1021/bi010713+] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Runx proteins constitute a family of mammalian transcription factors that interact with DNA through their evolutionarily conserved Runt domain. CBFbeta, alternatively denoted PEBP2beta, is the non-DNA-binding heterodimer partner and acts to enhance the DNA binding affinity of Runx proteins. Runx proteins and CBFbeta are associated with a variety of biological functions and human diseases; they are, for example, together the most frequent targets for chromosomal rearrangements in acute human leukemias. We have determined the solution structure and characterized the backbone dynamics of C-terminally truncated fragments containing residues 1-141 of CBFbeta. The present apo-CBFbeta structure is very similar to that seen in a Runt-CBFbeta complex. An evaluation of backbone (15)N NMR relaxation parameters shows that CBFbeta is a rigid molecule with high order parameters throughout the backbone; the only regions displaying significant dynamics are a long loop and the C-terminal alpha-helix. A few residues display relaxation behavior indicating conformational exchange on microsecond to millisecond time scales, but only one of these is located at the Runt binding surface. Our structure and dynamics analysis of CBFbeta therefore suggests that the protein binds to Runt without large conformational changes or induced folding ("lock-and-key" interaction). The apo-CBFbeta structure presented here exhibits several significant differences with two other published NMR ensembles of very similar protein fragments. The differences are located in four regions outside of the central beta-barrel, whereas the beta-barrel itself is almost identical in the three NMR structures. The comparison illustrates that independently determined NMR structures may display rather large differences in backbone conformation in regions that appear to be well-defined in each of the calculated NMR ensembles.
Collapse
Affiliation(s)
- M Wolf-Watz
- Department of Biotechnology, Center for Structural Biochemistry, Royal Institute of Technology (KTH), Novum, S-141 57 Huddinge, Sweden
| | | | | |
Collapse
|
40
|
Sudhakar S, Katz MS, Elango N. Analysis of type-I and type-II RUNX2 protein expression in osteoblasts. Biochem Biophys Res Commun 2001; 286:74-9. [PMID: 11485310 DOI: 10.1006/bbrc.2001.5363] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Runt-related transcription factor-2 (RUNX2) is expressed as two isoforms (type-I and type-II) differing only in their amino terminal sequences. The amino terminus of type-I contains MRIPV instead of MASNSLFSAVTPCQQSFFW in type-II. Although type-II mRNA has been considered osteoblast specific, the RUNX2 protein isoforms expressed in osteoblasts have not yet been identified. Using antisera generated against the two different amino terminal sequences of type-I and type-II RUNX2, we show the expression of both isoforms in cells with the mature osteoblast phenotype (fetal rat calvarial cells, and ROS 17/2.8, SaOS-2 and U2OS osteosarcoma cell lines), but only type-I in partially differentiated osteoblast-like cells (the UMR-106 osteosarcoma cell line). Since UMR-106 cells express both type-I and type-II mRNAs, our results suggest that the translation of type-II mRNA is repressed in these cells. No RUNX1 and RUNX3 proteins are detected in any of the osteoblastic cells tested. The antisera we have generated will be useful for studies relating expression of RUNX2 isoforms to control of osteoblast differentiation.
Collapse
Affiliation(s)
- S Sudhakar
- Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, University of Texas Health Science Center at San Antonio, San Antonio, TX 78284, USA
| | | | | |
Collapse
|
41
|
Choi JY, Pratap J, Javed A, Zaidi SK, Xing L, Balint E, Dalamangas S, Boyce B, van Wijnen AJ, Lian JB, Stein JL, Jones SN, Stein GS. Subnuclear targeting of Runx/Cbfa/AML factors is essential for tissue-specific differentiation during embryonic development. Proc Natl Acad Sci U S A 2001; 98:8650-5. [PMID: 11438701 PMCID: PMC37490 DOI: 10.1073/pnas.151236498] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Runx (Cbfa/AML) transcription factors are critical for tissue-specific gene expression. A unique targeting signal in the C terminus directs Runx factors to discrete foci within the nucleus. Using Runx2/CBFA1/AML3 and its essential role in osteogenesis as a model, we investigated the fundamental importance of fidelity of subnuclear localization for tissue differentiating activity by deleting the intranuclear targeting signal via homologous recombination. Mice homozygous for the deletion (Runx2 Delta C) do not form bone due to maturational arrest of osteoblasts. Heterozygotes do not develop clavicles, but are otherwise normal. These phenotypes are indistinguishable from those of the homozygous and heterozygous null mutants, indicating that the intranuclear targeting signal is a critical determinant for function. The expressed truncated Runx2 Delta C protein enters the nucleus and retains normal DNA binding activity, but shows complete loss of intranuclear targeting. These results demonstrate that the multifunctional N-terminal region of the Runx2 protein is not sufficient for biological activity. We conclude that subnuclear localization of Runx factors in specific foci together with associated regulatory functions is essential for control of Runx-dependent genes involved in tissue differentiation during embryonic development.
Collapse
Affiliation(s)
- J Y Choi
- Department of Cell Biology, University of Massachusetts Medical School, 55 Lake Avenue, North Worcester, MA 01655, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Nagata T, Werner MH. Functional mutagenesis of AML1/RUNX1 and PEBP2 beta/CBF beta define distinct, non-overlapping sites for DNA recognition and heterodimerization by the Runt domain. J Mol Biol 2001; 308:191-203. [PMID: 11327761 DOI: 10.1006/jmbi.2001.4596] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Runt domain family of transcription factors play key roles in transcriptional regulation of definitive hematopoiesis and osteogenesis. This transcription factor family is characterized by a DNA-binding alpha-subunit harboring the Runt domain and a secondary subunit, beta, which binds to the Runt domain and enhances its interaction with DNA. Missense mutations in the Runt domain from either the blood or bone-related gene product are associated with the onset of acute human leukemia as well as a disease of skeletal patterning known as cleidocranial dysplasia. NMR "footprinting" analysis of Runt domain/beta/DNA ternary complexes in solution previously identified the likely residues that form the heterodimerization and DNA-binding surfaces of the Runt domain. Functional mutagenesis at 37 positions in the Runt domain or beta confirms the original identification of these interaction surfaces and reveals that the heterodimerization and DNA-binding surfaces of the Runt domain occur at distinct, non-overlapping sites within the domain. The analysis of an additional 21 disease-related missense mutations identified from patients with either blood or bone disease demonstrates that the primary defect in these patients is a failure in DNA-recognition by the Runt domain. The molecular basis for the DNA-binding defect is analyzed in the context of the three-dimensional structure of the Runt domain in binary and ternary protein/DNA complexes.
Collapse
Affiliation(s)
- T Nagata
- The Rockefeller University, 1230 York Avenue, Box 42, New York, NY 10021, USA
| | | |
Collapse
|
43
|
Miller JD, Stacy T, Liu PP, Speck NA. Core-binding factor beta (CBFbeta), but not CBFbeta-smooth muscle myosin heavy chain, rescues definitive hematopoiesis in CBFbeta-deficient embryonic stem cells. Blood 2001; 97:2248-56. [PMID: 11290585 DOI: 10.1182/blood.v97.8.2248] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Core-binding factor beta (CBFbeta) is the non-DNA-binding subunit of the heterodimeric CBFs. Genes encoding CBFbeta (CBFB), and one of the DNA-binding CBFalpha subunits, Runx1 (also known as CBFalpha2, AML1, and PEBP2alphaB), are required for normal hematopoiesis and are also frequent targets of chromosomal translocations in acute leukemias in humans. Homozygous disruption of either the Runx1 or Cbfb gene in mice results in embryonic lethality at midgestation due to hemorrhaging in the central nervous system, and severely impairs fetal liver hematopoiesis. Results of this study show that Cbfb-deficient mouse embryonic stem (ES) cells can differentiate into primitive erythroid colonies in vitro, but are impaired in their ability to produce definitive erythroid and myeloid colonies, mimicking the in vivo defect. Definitive hematopoiesis is restored by ectopic expression of full-length Cbfb transgenes, as well as by a transgene encoding only the heterodimerization domain of CBFbeta. In contrast, the CBFbeta-smooth muscle myosin heavy chain (SMMHC) fusion protein generated by the inv(16) associated with acute myeloid leukemias (M4Eo) cannot rescue definitive hematopoiesis by Cbfb-deficient ES cells. Sequences responsible for the inability of CBFbeta-SMMHC to rescue definitive hematopoiesis reside in the SMMHC portion of the fusion protein. Results also show that the CBFbeta-SMMHC fusion protein transdominantly inhibits definitive hematopoiesis, but not to the same extent as homozygous loss of Runx1 or Cbfb. CBFbeta-SMMHC preferentially inhibits the differentiation of myeloid lineage cells, while increasing the number of blastlike cells in culture.
Collapse
Affiliation(s)
- J D Miller
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755, USA
| | | | | | | |
Collapse
|
44
|
Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, Shiina M, Sato K, Kumasaka T, Yamamoto M, Ishii S, Ogata K. Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta. Cell 2001; 104:755-67. [PMID: 11257229 DOI: 10.1016/s0092-8674(01)00271-9] [Citation(s) in RCA: 262] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The core binding factor (CBF) heterodimeric transcription factors comprised of AML/CBFA/PEBP2alpha/Runx and CBFbeta/PEBP2beta subunits are essential for differentiation of hematopoietic and bone cells, and their mutation is intimately related to the development of acute leukemias and cleidocranial dysplasia. Here, we present the crystal structures of the AML1/Runx-1/CBFalpha(Runt domain)-CBFbeta(core domain)-C/EBPbeta(bZip)-DNA, AML1/Runx-1/CBFalpha(Runt domain)-C/EBPbeta(bZip)-DNA, and AML1/Runx-1/CBFalpha(Runt domain)-DNA complexes. The hydrogen bonding network formed among CBFalpha(Runt domain) and CBFbeta, and CBFalpha(Runt domain) and DNA revealed the allosteric regulation mechanism of CBFalpha(Runt domain)-DNA binding by CBFbeta. The point mutations of CBFalpha related to the aforementioned diseases were also mapped and their effect on DNA binding is discussed.
Collapse
Affiliation(s)
- T H Tahirov
- Kanagawa Academy of Science and Technology (KAST), Yokohama City University School of Medicine, 3-9 Fukuura, Yokohama 236-0004, Kanazawa-ku, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Huang G, Shigesada K, Ito K, Wee HJ, Yokomizo T, Ito Y. Dimerization with PEBP2beta protects RUNX1/AML1 from ubiquitin-proteasome-mediated degradation. EMBO J 2001; 20:723-33. [PMID: 11179217 PMCID: PMC145428 DOI: 10.1093/emboj/20.4.723] [Citation(s) in RCA: 227] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The RUNX family genes are the mammalian homologs of the Drosophila genes runt and lozenge, and members of this family function as master regulators of definitive hematopoiesis and osteogenesis. The RUNX genes encode the alpha subunit of the transcription factor PEBP2/CBF. The beta subunit consists of the non-RUNX protein PEBP2beta. We found that RUNX1/AML1, which is essential for hematopoiesis, is continuously subjected to proteolytic degradation mediated by the ubiquitin-proteasome pathway. When PEBP2beta is present, however, the ubiquitylation of RUNX1 is abrogated and this causes a dramatic inhibition of RUNX1 proteolysis. Heterodimerization between PEBP2beta and RUNX1 thus appears to be an essential step in the generation of transcriptionally competent RUNX1. Consistent with this notion, RUNX1 was barely detected in PEBP2beta(-/-) mouse. CBF(PEBP2)beta- SMMHC, the chimeric protein associated with inv(16) acute myeloid leukemia, was found to protect RUNX1 from proteolytic degradation more efficiently than PEBP2beta. These results reveal a hitherto unknown and major role of PEBP2beta, namely that it regulates RUNX1 by controlling its turnover. This has allowed us to gain new insights into the mechanism of leukemogenesis by CBFbeta-SMMHC.
Collapse
Affiliation(s)
| | - Katsuya Shigesada
- Laboratory of Cell Regulation, Department of Viral Oncology, and
Molecular Genetics, Institute for Virus Research, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan Corresponding author e-mail:
| | | | | | | | - Yoshiaki Ito
- Laboratory of Cell Regulation, Department of Viral Oncology, and
Molecular Genetics, Institute for Virus Research, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan Corresponding author e-mail:
| |
Collapse
|
46
|
Wolf-Watz M, Bäckström S, Grundström T, Sauer U, Härd T. Chloride binding by the AML1/Runx1 transcription factor studied by NMR. FEBS Lett 2001; 488:81-4. [PMID: 11163800 DOI: 10.1016/s0014-5793(00)02390-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It is known that the DNA binding Runt domain of the AML1/Runx1 transcription factor coordinates Cl(-) ions. In this paper we have determined Cl(-) binding affinities of AML1 by (35)Cl nuclear magnetic resonance (NMR) linewidth analysis. The Runt domain binds Cl(-) with a dissociation constant (K(d,Cl)) of 34 mM. If CBFbeta is added to form a 1:1 complex, the K(d,Cl) value increases to 56 mM. Homology modeling suggests that a high occupancy Cl(-) binding site overlaps with the DNA binding surface. NMR data show that DNA displaces this Cl(-) ion. Possible biological roles of Cl(-) binding are discussed based on these findings.
Collapse
Affiliation(s)
- M Wolf-Watz
- Department of Biotechnology, Royal Institute of Technology, Center for Structural Biochemistry, Novum, Huddinge, Sweden
| | | | | | | | | |
Collapse
|
47
|
Tang YY, Shi J, Zhang L, Davis A, Bravo J, Warren AJ, Speck NA, Bushweller JH. Energetic and functional contribution of residues in the core binding factor beta (CBFbeta ) subunit to heterodimerization with CBFalpha. J Biol Chem 2000; 275:39579-88. [PMID: 10984496 DOI: 10.1074/jbc.m007350200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Core-binding factors (CBFs) are a small family of heterodimeric transcription factors that play critical roles in several developmental pathways, including hematopoiesis and bone development. Mutations in CBF genes are found in leukemias and bone disorders. CBFs consist of a DNA-binding CBFalpha subunit (Runx1, Runx2, or Runx3) and a non-DNA-binding CBFbeta subunit. CBFalpha binds DNA in a sequence-specific manner, whereas CBFbeta enhances DNA binding by CBFalpha. Recent structural analyses of the DNA-binding Runt domain of CBFalpha and the CBFbeta subunit identified the heterodimerization surfaces on each subunit. Here we identify amino acids in CBFbeta that mediate binding to CBFalpha. We determine the energy contributed by each of these amino acids to heterodimerization and the importance of these residues for in vivo function. These data refine the structural analyses and further support the hypothesis that CBFbeta enhances DNA binding by inducing a conformational change in the Runt domain.
Collapse
Affiliation(s)
- Y Y Tang
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zebrafish homolog of the leukemia gene CBFB: its expression during embryogenesis and its relationship to scland gata-1 in hematopoiesis. Blood 2000. [DOI: 10.1182/blood.v96.13.4178] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Abstract
Mammalian CBFB encodes a transcription factor (CBFβ) that in combination with CBFα2 binds to specific DNA sequences and regulates expression of a number of hematopoietic genes.CBFB is associated with human leukemias through a chromosome 16 inversion and is essential for definitive hematopoiesis during mouse embryo development. We have isolated a zebrafishcbfb complementary DNA (cDNA) clone from a zebrafish kidney cDNA library. This cbfb is highly homologous to human and mouseCBFB/Cbfb genes at both the DNA and protein level. In biochemical analyses, cbfβ binds to human CBFα2 and enhances its DNA binding. During zebrafish development, cbfb is expressed in the lateral plate mesoderm at tail bud stage and in the intermediate cell mass (ICM, the location of embryonic hematopoiesis) between the 21- to 26-somite stages. The cbfb is also expressed in Rohon-Beard cells, cranial nerve ganglia, hindbrain, retina, branchial arches, jaw, and fin buds. Expression ofcbfb is decreased or absent in the ICM and Rohon-Beard cells in some hematopoietic mutants and is unaffected in others. We have also analyzed the expression of scl andgata-1 in the same hematopoietic mutants to ascertain the relative order of these transcription factors to cbfb in zebrafish hematopoiesis. Our results indicate that cbfb is expressed in early hematopoietic progenitors and that its expression pattern in the hematopoietic mutants is similar to that ofscl.
Collapse
|
49
|
Zebrafish homolog of the leukemia gene CBFB: its expression during embryogenesis and its relationship to scland gata-1 in hematopoiesis. Blood 2000. [DOI: 10.1182/blood.v96.13.4178.h8004178_4178_4184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mammalian CBFB encodes a transcription factor (CBFβ) that in combination with CBFα2 binds to specific DNA sequences and regulates expression of a number of hematopoietic genes.CBFB is associated with human leukemias through a chromosome 16 inversion and is essential for definitive hematopoiesis during mouse embryo development. We have isolated a zebrafishcbfb complementary DNA (cDNA) clone from a zebrafish kidney cDNA library. This cbfb is highly homologous to human and mouseCBFB/Cbfb genes at both the DNA and protein level. In biochemical analyses, cbfβ binds to human CBFα2 and enhances its DNA binding. During zebrafish development, cbfb is expressed in the lateral plate mesoderm at tail bud stage and in the intermediate cell mass (ICM, the location of embryonic hematopoiesis) between the 21- to 26-somite stages. The cbfb is also expressed in Rohon-Beard cells, cranial nerve ganglia, hindbrain, retina, branchial arches, jaw, and fin buds. Expression ofcbfb is decreased or absent in the ICM and Rohon-Beard cells in some hematopoietic mutants and is unaffected in others. We have also analyzed the expression of scl andgata-1 in the same hematopoietic mutants to ascertain the relative order of these transcription factors to cbfb in zebrafish hematopoiesis. Our results indicate that cbfb is expressed in early hematopoietic progenitors and that its expression pattern in the hematopoietic mutants is similar to that ofscl.
Collapse
|
50
|
High incidence of biallelic point mutations in the Runt domain of the AML1/PEBP2αB gene in Mo acute myeloid leukemia and in myeloid malignancies with acquired trisomy 21. Blood 2000. [DOI: 10.1182/blood.v96.8.2862.h8002862_2862_2869] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The AML1 gene, situated in 21q22, is often rearranged in acute leukemias through t(8;21) translocation, t(12;21) translocation, or less often t(3;21) translocation. Recently, point mutations in the Runt domain of the AML1 gene have also been reported in leukemia patients. Observations for mutations of the Runt domain of the AML1 gene in bone marrow cells were made in 300 patients, including 131 with acute myeloid leukemia (AML), 94 with myelodysplastic syndrome (MDS), 28 with blast crisis chronic myeloid leukemia (CML), 3 with atypical CML, 41 with acute lymphoblastic leukemia (ALL), and 3 with essential thrombocythemia (ET). Forty-one of the patients had chromosome 21 abnormalities, including t(8;21) in 6 of the patients with AML, t(12;21) in 8 patients with ALL, acquired trisomy 21 in 17 patients, tetrasomy 21 in 7 patients, and constitutional trisomy 21 (Down syndrome) in 3 patients. A point mutation was found in 14 cases (4.7%), including 9 (22%) of the 41 patients with AML of the Mo type (MoAML) (none of them had detectable chromosome 21 rearrangement) and 5 (38%) of the 13 myeloid malignancies with acquired trisomy 21 (1 M1AML, 2 M2AML, 1 ET, and 1 atypical CML). In at least 8 of 9 mutated cases of MoAML, both AML alleles were mutated: 3 patients had different stop codon mutations of the 2 AML1 alleles, and 5 patients had the same missense or stop codon mutation in both AML1 alleles, which resulted in at least 3 of the patients having duplication of the mutated allele and deletion of the normal residual allele, as shown by FISH analysis and by comparing microsatellite analyses of several chromosome 21 markers on diagnosis and remission samples. In the remaining mutated cases, with acquired trisomy 21, a missense mutation of AML1, which involved 2 of the 3 copies of the AML1 gene, was found. Four of the 7 mutated cases could be reanalyzed in complete remission, and no AML1 mutation was found, showing that mutations were acquired in the leukemic clone. In conclusion, these findings confirm the possibility of mutations of the Runt domain of the AML1 gene in leukemias, mainly in MoAML and in myeloid malignancies with acquired trisomy 21. AML1 mutations, in MoAML, involved both alleles and probably lead to nonfunctional AML1 protein. As AML1 protein regulates the expression of the myeloperoxidase gene, the relationship between AML1 mutations and Mo phenotype in AML will have to be further explored.
Collapse
|