1
|
Li M, Dai M, Cheng B, Li S, Guo E, Fu J, Ma T, Yu B. Strategies that regulate LSD1 for novel therapeutics. Acta Pharm Sin B 2024; 14:1494-1507. [PMID: 38572094 PMCID: PMC10985039 DOI: 10.1016/j.apsb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Histone methylation plays crucial roles in regulating chromatin structure and gene transcription in epigenetic modifications. Lysine-specific demethylase 1 (LSD1), the first identified histone demethylase, is universally overexpressed in various diseases. LSD1 dysregulation is closely associated with cancer, viral infections, and neurodegenerative diseases, etc., making it a promising therapeutic target. Several LSD1 inhibitors and two small-molecule degraders (UM171 and BEA-17) have entered the clinical stage. LSD1 can remove methyl groups from histone 3 at lysine 4 or lysine 9 (H3K4 or H3K9), resulting in either transcription repression or activation. While the roles of LSD1 in transcriptional regulation are well-established, studies have revealed that LSD1 can also be dynamically regulated by other factors. For example, the expression or activity of LSD1 can be regulated by many proteins that form transcriptional corepressor complexes with LSD1. Moreover, some post-transcriptional modifications and cellular metabolites can also regulate LSD1 expression or its demethylase activity. Therefore, in this review, we will systematically summarize how proteins involved in the transcriptional corepressor complex, various post-translational modifications, and metabolites act as regulatory factors for LSD1 activity.
Collapse
Affiliation(s)
- Meng Li
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Mengge Dai
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Bing Cheng
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Shaotong Li
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Enhui Guo
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Junwei Fu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Ma
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
| | - Bin Yu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450000, China
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
2
|
Ainciburu M, Ezponda T, Berastegui N, Alfonso-Pierola A, Vilas-Zornoza A, San Martin-Uriz P, Alignani D, Lamo-Espinosa J, San-Julian M, Jiménez-Solas T, Lopez F, Muntion S, Sanchez-Guijo F, Molero A, Montoro J, Serrano G, Diaz-Mazkiaran A, Lasaga M, Gomez-Cabrero D, Diez-Campelo M, Valcarcel D, Hernaez M, Romero JP, Prosper F. Uncovering perturbations in human hematopoiesis associated with healthy aging and myeloid malignancies at single-cell resolution. eLife 2023; 12:79363. [PMID: 36629404 PMCID: PMC9904760 DOI: 10.7554/elife.79363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Early hematopoiesis is a continuous process in which hematopoietic stem and progenitor cells (HSPCs) gradually differentiate toward specific lineages. Aging and myeloid malignant transformation are characterized by changes in the composition and regulation of HSPCs. In this study, we used single-cell RNA sequencing (scRNA-seq) to characterize an enriched population of human HSPCs obtained from young and elderly healthy individuals. Based on their transcriptional profile, we identified changes in the proportions of progenitor compartments during aging, and differences in their functionality, as evidenced by gene set enrichment analysis. Trajectory inference revealed that altered gene expression dynamics accompanied cell differentiation, which could explain aging-associated changes in hematopoiesis. Next, we focused on key regulators of transcription by constructing gene regulatory networks (GRNs) and detected regulons that were specifically active in elderly individuals. Using previous findings in healthy cells as a reference, we analyzed scRNA-seq data obtained from patients with myelodysplastic syndrome (MDS) and detected specific alterations of the expression dynamics of genes involved in erythroid differentiation in all patients with MDS such as TRIB2. In addition, the comparison between transcriptional programs and GRNs regulating normal HSPCs and MDS HSPCs allowed identification of regulons that were specifically active in MDS cases such as SMAD1, HOXA6, POU2F2, and RUNX1 suggesting a role of these transcription factors (TFs) in the pathogenesis of the disease. In summary, we demonstrate that the combination of single-cell technologies with computational analysis tools enable the study of a variety of cellular mechanisms involved in complex biological systems such as early hematopoiesis and can be used to dissect perturbed differentiation trajectories associated with perturbations such as aging and malignant transformation. Furthermore, the identification of abnormal regulatory mechanisms associated with myeloid malignancies could be exploited for personalized therapeutic approaches in individual patients.
Collapse
Affiliation(s)
- Marina Ainciburu
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Teresa Ezponda
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Nerea Berastegui
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
| | - Ana Alfonso-Pierola
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Clinica Universidad de NavarraPamplonaSpain
| | - Amaia Vilas-Zornoza
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Patxi San Martin-Uriz
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Diego Alignani
- Flow Cytometry Core, Universidad de NavarraPamplonaSpain
| | | | | | | | - Felix Lopez
- Hospital Universitario de SalamancaSalamancaSpain
| | - Sandra Muntion
- Hospital Universitario de SalamancaSalamancaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| | - Fermin Sanchez-Guijo
- Hospital Universitario de SalamancaSalamancaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| | - Antonieta Molero
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | - Julia Montoro
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | | | - Aintzane Diaz-Mazkiaran
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Computational Biology Program, Universidad de NavarraPamplonaSpain
| | - Miren Lasaga
- Translational Bioinformatics Unit, NavarraBiomedPamplonaSpain
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, NavarraBiomedPamplonaSpain
- Biological & Environmental Sciences & Engineering Division, King Abdullah University of Science and TechnologyThuwalSaudi Arabia
| | | | - David Valcarcel
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | - Mikel Hernaez
- Computational Biology Program, Universidad de NavarraPamplonaSpain
| | - Juan P Romero
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Felipe Prosper
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Clinica Universidad de NavarraPamplonaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| |
Collapse
|
3
|
Maiques-Diaz A, Nicosia L, Basma NJ, Romero-Camarero I, Camera F, Spencer GJ, Amaral FMR, Simeoni F, Wingelhofer B, Williamson AJK, Pierce A, Whetton AD, Somervaille TCP. HMG20B stabilizes association of LSD1 with GFI1 on chromatin to confer transcription repression and leukemia cell differentiation block. Oncogene 2022; 41:4841-4854. [PMID: 36171271 PMCID: PMC7613766 DOI: 10.1038/s41388-022-02471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/09/2022]
Abstract
Pharmacologic inhibition of LSD1 induces molecular and morphologic differentiation of blast cells in acute myeloid leukemia (AML) patients harboring MLL gene translocations. In addition to its demethylase activity, LSD1 has a critical scaffolding function at genomic sites occupied by the SNAG domain transcription repressor GFI1. Importantly, inhibitors block both enzymatic and scaffolding activities, in the latter case by disrupting the protein:protein interaction of GFI1 with LSD1. To explore the wider consequences of LSD1 inhibition on the LSD1 protein complex we applied mass spectrometry technologies. We discovered that the interaction of the HMG-box protein HMG20B with LSD1 was also disrupted by LSD1 inhibition. Downstream investigations revealed that HMG20B is co-located on chromatin with GFI1 and LSD1 genome-wide; the strongest HMG20B binding co-locates with the strongest GFI1 and LSD1 binding. Functional assays demonstrated that HMG20B depletion induces leukemia cell differentiation and further revealed that HMG20B is required for the transcription repressor activity of GFI1 through stabilizing LSD1 on chromatin at GFI1 binding sites. Interaction of HMG20B with LSD1 is through its coiled-coil domain. Thus, HMG20B is a critical component of the GFI1:LSD1 transcription repressor complex which contributes to leukemia cell differentiation block.
Collapse
Affiliation(s)
- Alba Maiques-Diaz
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Luciano Nicosia
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Naseer J Basma
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Isabel Romero-Camarero
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Francesco Camera
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Gary J Spencer
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Fabio M R Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Fabrizio Simeoni
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Bettina Wingelhofer
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Andrew J K Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, 27 Palatine Road, Manchester, M20 3LJ, UK
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, 27 Palatine Road, Manchester, M20 3LJ, UK
- School of Medical and Health Sciences, College of Human Sciences, Fron Heulog Bangor University, Bangor, LL57 2TH, UK
| | - Anthony D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, 27 Palatine Road, Manchester, M20 3LJ, UK
- School of Veterinary Medicine and School of Biosciences and Medicine, University of Surrey, VSM Building, University of Surrey, Guildford, GU2 7AL, UK
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre Building, 555 Wilmslow Road, Manchester, M20 4GJ, UK.
| |
Collapse
|
4
|
Cheng T, Li X, Chen J, Yang L, Liu J, Song G, Ma H. Investigation of hub genes involved in Turner syndrome using biological informatics methods. Medicine (Baltimore) 2022; 101:e29069. [PMID: 35356930 PMCID: PMC10684194 DOI: 10.1097/md.0000000000029069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 02/25/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND This study aimed to explore candidate genes and their potential interaction mechanism critical to the pathophysiology of Turner syndrome by using the Gene Expression Omnibus database. METHODS GSE58435 data set was obtained by querying the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened using R and subsequently annotated by Gene Ontology. Functional enrichment analysis was performed based on the Kyoto Encyclopedia of Genes and Genomes database for annotation, visualization, and integrated discovery. A protein-protein interaction network of different genes was constructed based on the STRING database, in which hub genes were explored through Cytoscape software. The expression of the hub genes was verified by analyzing the gene expression in the GSE46687 data set. RESULTS A total of 733 differential genes were identified. These differentially expressed genes were significantly enriched in nucleoplasm and nucleus. Their molecular function was concentrated on DNA binding and transcription, coronary artery, and adipose tissue development. According to the annotation of Kyoto Encyclopedia of Genes and Genomes, the identified DEGs were mainly enriched in inflammatory mediator regulation of TRP channels, osteoclast differentiation. A total of 10 hub genes (HIST1H2BA, TRIM71, HIST1H2BB, HIST1H4D, TNF, TP53BP1, CDCA8, EGF, HMG20B, and BCL9) were identified from the constructed protein-protein interaction network. These genes were discovered to be highly expressed in osteoclasts, ovaries, digestive tract, blood, and lymphatic tissues through the online application of human protein atlas. CONCLUSION In this study, 733 DEGs and 10 hub genes were identified. They would be new candidate targets in Turner syndrome.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huijuan Ma
- Correspondence: Huijuan Ma, Hebei General Hospital, Shijiazhuang, Hebei, China (e-mail: ).
| |
Collapse
|
5
|
Ulyanova T, Cherone JM, Sova P, Papayannopoulou T. α4-Integrin deficiency in human CD34+ cells engenders precocious erythroid differentiation but inhibits enucleation. Exp Hematol 2022; 108:16-25. [DOI: 10.1016/j.exphem.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/26/2022]
|
6
|
Fu Y, Cai L, Lei X, Wang D. Circular RNA ABCB10 promotes hepatocellular carcinoma progression by increasing HMG20A expression by sponging miR-670-3p. Cancer Cell Int 2019; 19:338. [PMID: 31889891 PMCID: PMC6915995 DOI: 10.1186/s12935-019-1055-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Background/aims The dysregulation of circABCB10 may play an critical role in tumor progression. However, its function in liver cancer (HCC) is still unclear. Therefore, this experimental design is based on circABCB10 to explore the pathogenesis of HCC. Methods The expression of circABCB10 and miR-670-3p in HCC tissues was detected by RT-qPCR. CCK-8, Brdu incorporation, colony formation and transwell assays were used to determine the effect of circABCB10 on HCC cell proliferation and migration. Target gene prediction and screening, luciferase reporter assays were used to validate downstream target genes of circABCB10 and miR-670-3p. HMG20A expression was detected by RT-qPCR and Western blotting. The tumor changes in mice were detected by in nude mice. Results CircABCB10 was significantly increased in HCC tissues and cell lines, and high CircABCB10 expression was directly associated with low survival in HCC patients. Silencing of circABCB10 inhibited proliferation and invasion of hepatocellular carcinoma. In addition, circABCB10 acted as a sponge of miR-670-3p to upregulate HMG20A expression. In addition, overexpression of miR-670-3p or knockdown of HMG20A reversed the carcinogenic effects of circABCB10 in HCC. There was a negative correlation between the expression of circABCB10 and miR-670-3p, and a positive correlation between the expression of circABCB10 and HMG20A in HCC tissues. Conclusion circABCB10 promoted HCC progression by modulating the miR-670-3p/HMG20A axis, and circABCB10 may be a potential therapeutic target for HCC. Trail registration JL1H384739, registered at Sep 09, 2014.
Collapse
Affiliation(s)
- Yu Fu
- 1Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, 130021 Jilin People's Republic of China
| | - Limin Cai
- 2Department of Anesthesiology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021 Jilin People's Republic of China
| | - Xuexue Lei
- 1Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, 130021 Jilin People's Republic of China
| | - Dunwei Wang
- 2Department of Anesthesiology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021 Jilin People's Republic of China
| |
Collapse
|
7
|
Molecular Modelling of Islet β-Cell Adaptation to Inflammation in Pregnancy and Gestational Diabetes Mellitus. Int J Mol Sci 2019; 20:ijms20246171. [PMID: 31817798 PMCID: PMC6941051 DOI: 10.3390/ijms20246171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022] Open
Abstract
Gestational diabetes mellitus (GDM), a metabolic disease that develops with the increase in insulin resistance during late pregnancy, is currently one of the most common complications affecting pregnancy. The polygenic nature of GDM, together with the interplay between different genetic variants with nutritional and environmental factors has hindered the full understanding of the etiology of this disease. However, an important genetic overlap has been found with type 2 diabetes mellitus (T2DM) and, as in the case of T2DM, most of the identified loci are associated with β-cell function. Early detection of GDM and adequate interventions to control the maternal glycemia are necessary to avoid the adverse outcomes for both the mother and the offspring. The in utero exposure to the diabetic milieu predispose these children for future diseases, among them T2DM, originating a vicious circle implicated in the increased prevalence of both GDM and T2DM. The involvement of inflammatory processes in the development of GDM highlights the importance of pancreatic β-cell factors able to favor the adaptation processes required during gestation, concomitantly with the protection of the islets from an inflammatory milieu. In this regard, two members of the Pax family of transcription factors, PAX4 and PAX8, together with the chromatin remodeler factor HMG20A, have gained great relevance due to their involvement in β-cell mass adaptation together with their anti-inflammatory properties. Mutations in these factors have been associated with GDM, highlighting these as novel candidates for genetic screening analysis in the identification of women at risk of developing GDM.
Collapse
|
8
|
van Bergen MGJM, van der Reijden BA. Targeting the GFI1/1B-CoREST Complex in Acute Myeloid Leukemia. Front Oncol 2019; 9:1027. [PMID: 31649884 PMCID: PMC6794713 DOI: 10.3389/fonc.2019.01027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/23/2019] [Indexed: 11/21/2022] Open
Abstract
One of the hallmarks of acute myeloid leukemia (AML) is a block in cellular differentiation. Recent studies have shown that small molecules targeting Lysine Specific Demethylase 1A (KDM1A) may force the malignant cells to terminally differentiate. KDM1A is a core component of the chromatin binding CoREST complex. Together with histone deacetylases CoREST regulates gene expression by histone 3 demethylation and deacetylation. The transcription factors GFI1 and GFI1B (for growth factor independence) are major interaction partners of KDM1A and recruit the CoREST complex to chromatin in myeloid cells. Recent studies show that the small molecules that target KDM1A disrupt the GFI1/1B-CoREST interaction and that this is key to inducing terminal differentiation of leukemia cells.
Collapse
Affiliation(s)
| | - Bert A. van der Reijden
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
9
|
McClellan D, Casey MJ, Bareyan D, Lucente H, Ours C, Velinder M, Singer J, Lone MD, Sun W, Coria Y, Mason CC, Engel ME. Growth Factor Independence 1B-Mediated Transcriptional Repression and Lineage Allocation Require Lysine-Specific Demethylase 1-Dependent Recruitment of the BHC Complex. Mol Cell Biol 2019; 39:e00020-19. [PMID: 30988160 PMCID: PMC6580704 DOI: 10.1128/mcb.00020-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/30/2019] [Accepted: 04/09/2019] [Indexed: 12/16/2022] Open
Abstract
Growth factor independence 1B (GFI1B) coordinates assembly of transcriptional repressor complexes comprised of corepressors and histone-modifying enzymes to control gene expression programs governing lineage allocation in hematopoiesis. Enforced expression of GFI1B in K562 erythroleukemia cells favors erythroid over megakaryocytic differentiation, providing a platform to define molecular determinants of binary fate decisions triggered by GFI1B. We deployed proteome-wide proximity labeling to identify factors whose inclusion in GFI1B complexes depends upon GFI1B's obligate effector, lysine-specific demethylase 1 (LSD1). We show that GFI1B preferentially recruits core and putative elements of the BRAF-histone deacetylase (HDAC) (BHC) chromatin-remodeling complex (LSD1, RCOR1, HMG20A, HMG20B, HDAC1, HDAC2, PHF21A, GSE1, ZMYM2, and ZNF217) in an LSD1-dependent manner to control acquisition of erythroid traits by K562 cells. Among these elements, depletion of both HMG20A and HMG20B or of GSE1 blocks GFI1B-mediated erythroid differentiation, phenocopying impaired differentiation brought on by LSD1 depletion or disruption of GFI1B-LSD1 binding. These findings demonstrate the central role of the GFI1B-LSD1 interaction as a determinant of BHC complex recruitment to enable cell fate decisions driven by GFI1B.
Collapse
Affiliation(s)
- David McClellan
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mattie J Casey
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Diana Bareyan
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Helena Lucente
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Christopher Ours
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Matthew Velinder
- Department of Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jason Singer
- Department of Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mehraju Din Lone
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Wenxiang Sun
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Yunuen Coria
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Clinton C Mason
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Michael E Engel
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
- Center for Investigational Therapeutics, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Nuclear Control of Cell Growth and Differentiation Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Liu L, Wan X, Zhou P, Zhou X, Zhang W, Hui X, Yuan X, Ding X, Zhu R, Meng G, Xiao H, Ma F, Huang H, Song X, Zhou B, Xiong S, Zhang Y. The chromatin remodeling subunit Baf200 promotes normal hematopoiesis and inhibits leukemogenesis. J Hematol Oncol 2018; 11:27. [PMID: 29482581 PMCID: PMC5828314 DOI: 10.1186/s13045-018-0567-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/05/2018] [Indexed: 11/10/2022] Open
Abstract
Background Adenosine triphosphate (ATP)-dependent chromatin remodeling SWI/SNF-like BAF and PBAF complexes have been implicated in the regulation of stem cell function and cancers. Several subunits of BAF or PBAF, including BRG1, BAF53a, BAF45a, BAF180, and BAF250a, are known to be involved in hematopoiesis. Baf200, a subunit of PBAF complex, plays a pivotal role in heart morphogenesis and coronary artery angiogenesis. However, little is known on the importance of Baf200 in normal and malignant hematopoiesis. Methods Utilizing Tie2-Cre-, Vav-iCre-, and Mx1-Cre-mediated Baf200 gene deletion combined with fetal liver/bone marrow transplantation, we investigated the function of Baf200 in fetal and adult hematopoiesis. In addition, a mouse model of MLL-AF9-driven leukemogenesis was used to study the role of Baf200 in malignant hematopoiesis. We also explored the potential mechanism by using RNA-seq, RT-qPCR, cell cycle, and apoptosis assays. Results Tie2-Cre-mediated loss of Baf200 causes perinatal death due to defective erythropoiesis and impaired hematopoietic stem cell expansion in the fetal liver. Vav-iCre-mediated loss of Baf200 causes only mild anemia and enhanced extramedullary hematopoiesis. Fetal liver hematopoietic stem cells from Tie2-Cre+, Baf200f/f or Vav-iCre+, Baf200f/f embryos and bone marrow hematopoietic stem cells from Vav-iCre+, Baf200f/f mice exhibited impaired long-term reconstitution potential in vivo. A cell-autonomous requirement of Baf200 for hematopoietic stem cell function was confirmed utilizing the interferon-inducible Mx1-Cre mouse strain. Transcriptomes analysis revealed that expression of several erythropoiesis- and hematopoiesis-associated genes were regulated by Baf200. In addition, loss of Baf200 in a mouse model of MLL-AF9-driven leukemogenesis accelerates the tumor burden and shortens the host survival. Conclusion Our current studies uncover critical roles of Baf200 in both normal and malignant hematopoiesis and provide a potential therapeutic target for suppressing the progression of leukemia without interfering with normal hematopoiesis. Electronic supplementary material The online version of this article (10.1186/s13045-018-0567-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lulu Liu
- Institute of Biology and Medical Sciences, Soochow University, No. 199 Ren'ai Rd, Suzhou, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China
| | - Xiaoling Wan
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peipei Zhou
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyuan Zhou
- University of Chinese Academy of Sciences, Beijing, China.,CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Xinhui Hui
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiujie Yuan
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaodan Ding
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ruihong Zhu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guangxun Meng
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hui Xiao
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Feng Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianmin Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Shanghai General Hospital, Shanghai, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.
| | - Sidong Xiong
- Institute of Biology and Medical Sciences, Soochow University, No. 199 Ren'ai Rd, Suzhou, China.
| | - Yan Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
11
|
Mayer RL, Schwarzmeier JD, Gerner MC, Bileck A, Mader JC, Meier-Menches SM, Gerner SM, Schmetterer KG, Pukrop T, Reichle A, Slany A, Gerner C. Proteomics and metabolomics identify molecular mechanisms of aging potentially predisposing for chronic lymphocytic leukemia. Mol Cell Proteomics 2017; 17:290-303. [PMID: 29196338 DOI: 10.1074/mcp.ra117.000425] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Indexed: 01/07/2023] Open
Abstract
B cell chronic lymphocytic leukemia (B-CLL), the most common type of leukemia in adults, is still essentially incurable despite the development of novel therapeutic strategies. This reflects the incomplete understanding of the pathophysiology of this disease. A comprehensive proteome analysis of primary human B-CLL cells and B cells from younger as well as elderly healthy donors was performed. For comparison, the chronic B cell leukemia cell line JVM-13 was also included. A principal component analysis comprising 6,945 proteins separated these four groups, placing B cells of aged-matched controls between those of young donors and B-CLL patients, while identifying JVM-13 as poorly related cells. Mass spectrometric proteomics data have been made fully accessible via ProteomeXchange with identifier PXD006570-PXD006572, PXD006576, PXD006578, and PXD006589-PXD006591. Remarkably, B cells from aged controls displayed significant regulation of proteins related to stress management in mitochondria and ROS stress such as DLAT, FIS1, and NDUFAB1, and DNA repair, including RAD9A, MGMT, and XPA. ROS levels were indeed found significantly increased in B cells but not in T cells or monocytes from aged individuals. These alterations may be relevant for tumorigenesis and were observed similarly in B-CLL cells. In B-CLL cells, some remarkable unique features like the loss of tumor suppressor molecules PNN and JARID2, the stress-related serotonin transporter SLC6A4, and high expression of ZNF207, CCDC88A, PIGR and ID3, otherwise associated with stem cell phenotype, were determined. Alterations of metabolic enzymes were another outstanding feature in comparison to normal B cells, indicating increased beta-oxidation of fatty acids and increased consumption of glutamine. Targeted metabolomics assays corroborated these results. The present findings identify a potential proteome signature for immune senescence in addition to previously unrecognized features of B-CLL cells and suggest that aging may be accompanied by cellular reprogramming functionally relevant for predisposing B cells to transform to B-CLL cells.
Collapse
Affiliation(s)
- Rupert L Mayer
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry
| | - Josef D Schwarzmeier
- §Karl Landsteiner Institute for Bioanalytical Oncology, Karl Landsteiner Society, Vienna, Austria
| | - Marlene C Gerner
- ¶Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Andrea Bileck
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry
| | - Johanna C Mader
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry
| | | | - Samuel M Gerner
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry
| | | | - Tobias Pukrop
- ‖Department of Internal Medicine III, Haematology & Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Albrecht Reichle
- ‖Department of Internal Medicine III, Haematology & Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Astrid Slany
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry
| | - Christopher Gerner
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, .,**Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Abstract
The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor β subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.
Collapse
|
13
|
Abstract
B-cell lymphoma 11A (BCL11A) downregulation in human primary adult erythroid progenitors results in elevated expression of fetal γ-globin. Recent reports showed that BCL11A expression is activated by KLF1, leading to γ-globin repression. To study regulation of erythropoiesis and globin expression by KLF1 and BCL11A in an in vivo model, we used mice carrying a human β-globin locus transgene with combinations of Klf1 knockout, Bcl11a floxed, and EpoR(Cre) knockin alleles. We found a higher percentage of reticulocytes in adult Klf1(wt/ko) mice and a mild compensated anemia in Bcl11a(cko/cko) mice. These phenotypes were more pronounced in compound Klf1(wt/ko)::Bcl11a(cko/cko) mice. Analysis of Klf1(wt/ko), Bcl11a(cko/cko), and Klf1(wt/ko)::Bcl11a(cko/cko) mutant embryos demonstrated increased expression of mouse embryonic globins during fetal development. Expression of human γ-globin remained high in Bcl11a(cko/cko) embryos during fetal development, and this was further augmented in Klf1(wt/ko)::Bcl11a(cko/cko) embryos. After birth, expression of human γ-globin and mouse embryonic globins decreased in Bcl11a(cko/cko) and Klf1(wt/ko)::Bcl11a(cko/cko) mice, but the levels remained much higher than those observed in control animals. Collectively, our data support an important role for the KLF1-BCL11A axis in erythroid maturation and developmental regulation of globin expression.
Collapse
|
14
|
Fanis P, Gillemans N, Aghajanirefah A, Pourfarzad F, Demmers J, Esteghamat F, Vadlamudi RK, Grosveld F, Philipsen S, van Dijk TB. Five friends of methylated chromatin target of protein-arginine-methyltransferase[prmt]-1 (chtop), a complex linking arginine methylation to desumoylation. Mol Cell Proteomics 2012; 11:1263-73. [PMID: 22872859 DOI: 10.1074/mcp.m112.017194] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chromatin target of Prmt1 (Chtop) is a vertebrate-specific chromatin-bound protein that plays an important role in transcriptional regulation. As its mechanism of action remains unclear, we identified Chtop-interacting proteins using a biotinylation-proteomics approach. Here we describe the identification and initial characterization of Five Friends of Methylated Chtop (5FMC). 5FMC is a nuclear complex that can only be recruited by Chtop when the latter is arginine-methylated by Prmt1. It consists of the co-activator Pelp1, the Sumo-specific protease Senp3, Wdr18, Tex10, and Las1L. Pelp1 functions as the core of 5FMC, as the other components become unstable in the absence of Pelp1. We show that recruitment of 5FMC to Zbp-89, a zinc-finger transcription factor, affects its sumoylation status and transactivation potential. Collectively, our data provide a mechanistic link between arginine methylation and (de)sumoylation in the control of transcriptional activity.
Collapse
Affiliation(s)
- Pavlos Fanis
- Department of Cell Biology, Erasmus MC, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|