1
|
Cho B, Kim J, Kim S, An S, Hwang Y, Kim Y, Kwon D, Kim J. Epigenetic Dynamics in Reprogramming to Dopaminergic Neurons for Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403105. [PMID: 39279468 PMCID: PMC11538697 DOI: 10.1002/advs.202403105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/28/2024] [Indexed: 09/18/2024]
Abstract
Direct lineage reprogramming into dopaminergic (DA) neurons holds great promise for the more effective production of DA neurons, offering potential therapeutic benefits for conditions such as Parkinson's disease. However, the reprogramming pathway for fully reprogrammed DA neurons remains largely unclear, resulting in immature and dead-end states with low efficiency. In this study, using single-cell RNA sequencing, the trajectory of reprogramming DA neurons at multiple time points, identifying a continuous pathway for their reprogramming is analyzed. It is identified that intermediate cell populations are crucial for resetting host cell fate during early DA neuronal reprogramming. Further, longitudinal dissection uncovered two distinct trajectories: one leading to successful reprogramming and the other to a dead end. Notably, Arid4b, a histone modifier, as a crucial regulator at this branch point, essential for the successful trajectory and acquisition of mature dopaminergic neuronal identity is identified. Consistently, overexpressing Arid4b in the DA neuronal reprogramming process increases the yield of iDA neurons and effectively reverses the disease phenotypes observed in the PD mouse brain. Thus, gaining insights into the cellular trajectory holds significant importance for devising regenerative medicine strategies, particularly in the context of addressing neurodegenerative disorders like Parkinson's disease.
Collapse
Affiliation(s)
- Byounggook Cho
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Junyeop Kim
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Sumin Kim
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Saemin An
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Yerim Hwang
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Yunkyung Kim
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Daeyeol Kwon
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell ReprogrammingDepartment of Chemistry and Biomedical EngineeringDongguk UniversitySeoul04620Republic of Korea
| |
Collapse
|
2
|
Alganatay C, Balbasi E, Tuncbag N, Sezginmert D, Terzi Cizmecioglu N. SETD3 regulates endoderm differentiation of mouse embryonic stem cells through canonical Wnt signaling pathway. FASEB J 2024; 38:e23463. [PMID: 38334393 DOI: 10.1096/fj.202301883r] [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: 09/15/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
With self-renewal and pluripotency features, embryonic stem cells (ESCs) provide an invaluable tool to investigate early cell fate decisions. Pluripotency exit and lineage commitment depend on precise regulation of gene expression that requires coordination between transcription (TF) and chromatin factors in response to various signaling pathways. SET domain-containing 3 (SETD3) is a methyltransferase that can modify histones in the nucleus and actin in the cytoplasm. Through an shRNA screen, we previously identified SETD3 as an important factor in the meso/endodermal lineage commitment of mouse ESCs (mESC). In this study, we identified SETD3-dependent transcriptomic changes during endoderm differentiation of mESCs using time-course RNA-seq analysis. We found that SETD3 is involved in the timely activation of the endoderm-related gene network. The canonical Wnt signaling pathway was one of the markedly altered signaling pathways in the absence of SETD3. The assessment of Wnt transcriptional activity revealed a significant reduction in Setd3-deleted (setd3∆) mESCs coincident with a decrease in the nuclear pool of the key TF β-catenin level, though no change was observed in its mRNA or total protein level. Furthermore, a proximity ligation assay (PLA) found an interaction between SETD3 and β-catenin. We were able to rescue the differentiation defect by stably re-expressing SETD3 or activating the canonical Wnt signaling pathway by changing mESC culture conditions. Our results suggest that alterations in the canonical Wnt pathway activity and subcellular localization of β-catenin might contribute to the endoderm differentiation defect of setd3∆ mESCs.
Collapse
Affiliation(s)
- Ceren Alganatay
- Faculty of Arts and Sciences, Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Emre Balbasi
- Faculty of Arts and Sciences, Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | | | - Dersu Sezginmert
- Faculty of Arts and Sciences, Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Nihal Terzi Cizmecioglu
- Faculty of Arts and Sciences, Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
3
|
Terzi Çizmecioglu N. Application of CRISPR/Cas9 Pooled Screening for a Non-immediate Readout Model. Methods Mol Biol 2024; 2849:87-116. [PMID: 38441725 DOI: 10.1007/7651_2024_529] [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] [Indexed: 07/20/2024]
Abstract
Linking phenotypes to genetic components has been an essential part of novel drug discovery, and screening methods have been widely employed to achieve such a goal. Screens can be conducted in either pooled or arrayed formats. Although arrayed screenings provide a better and cheaper alternative in small scale, the larger-scale screenings are conducted in pooled manner. With its adaptability to various models and conditions, CRISPR/Cas9 technology provides an invaluable alternative to classical and RNAi-based screening methods. Combined with high-throughput sequencing and bioinformatics, CRISPR-/Cas9-based pooled screening methods provide unbiased and robust data. In this protocol, we employed CRISPR-/Cas9-based pooled screening for a non-binary and non-immediate readout.
Collapse
|
4
|
Robert VJ, Caron M, Gely L, Adrait A, Pakulska V, Couté Y, Chevalier M, Riedel CG, Bedet C, Palladino F. SIN-3 acts in distinct complexes to regulate the germline transcriptional program in Caenorhabditis elegans. Development 2023; 150:dev201755. [PMID: 38771303 PMCID: PMC10617626 DOI: 10.1242/dev.201755] [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: 03/08/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023]
Abstract
The transcriptional co-regulator SIN3 influences gene expression through multiple interactions that include histone deacetylases. Haploinsufficiency and mutations in SIN3 are the underlying cause of Witteveen-Kolk syndrome and related intellectual disability and autism syndromes, emphasizing its key role in development. However, little is known about the diversity of its interactions and functions in developmental processes. Here, we show that loss of SIN-3, the single SIN3 homolog in Caenorhabditis elegans, results in maternal-effect sterility associated with de-regulation of the germline transcriptome, including de-silencing of X-linked genes. We identify at least two distinct SIN3 complexes containing specific histone deacetylases and show that they differentially contribute to fertility. Single-cell, single-molecule fluorescence in situ hybridization reveals that in sin-3 mutants the X chromosome becomes re-expressed prematurely and in a stochastic manner in individual germ cells, suggesting a role for SIN-3 in its silencing. Furthermore, we identify histone residues whose acetylation increases in the absence of SIN-3. Together, this work provides a powerful framework for the in vivo study of SIN3 and associated proteins.
Collapse
Affiliation(s)
- Valerie J. Robert
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, 69007 Lyon, France
| | - Matthieu Caron
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, 69007 Lyon, France
| | - Loic Gely
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, 69007 Lyon, France
| | - Annie Adrait
- Grenoble Alpes, CEA, Inserm, UA13 BGE, CNRS, CEA, FR2048, 38000 Grenoble, France
| | - Victoria Pakulska
- Grenoble Alpes, CEA, Inserm, UA13 BGE, CNRS, CEA, FR2048, 38000 Grenoble, France
| | - Yohann Couté
- Grenoble Alpes, CEA, Inserm, UA13 BGE, CNRS, CEA, FR2048, 38000 Grenoble, France
| | - Manon Chevalier
- Department of Biosciences and Nutrition, Karolinska Institutet, Blickagången 16, 14157 Huddinge, Sweden
| | - Christian G. Riedel
- Department of Biosciences and Nutrition, Karolinska Institutet, Blickagången 16, 14157 Huddinge, Sweden
| | - Cecile Bedet
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, 69007 Lyon, France
| | - Francesca Palladino
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, 69007 Lyon, France
| |
Collapse
|
5
|
Terzi Çizmecioğlu N. ARID4B loss leads to activated STAT1-dependent interferon pathway in mouse embryonic stem cells and during meso/endodermal differentiation. J Turk Ger Gynecol Assoc 2023; 24:187-196. [PMID: 37675520 PMCID: PMC10493817 DOI: 10.4274/jtgga.galenos.2023.2023-7-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/26/2023] [Indexed: 09/08/2023] Open
Abstract
Objective Proper deactivation of the pluripotency network and activation of a lineage-specific gene expression program are critical for mouse embryonic stem cell (mESC) differentiation. This is achieved by the coordinated action of transcription and chromatin factors. Our previous work identified ARID4B as a critical chromatin factor for mesoderm and endoderm differentiation. As part of a histone deacetylase complex, ARID4B plays a role in transcriptional suppression of its direct targets. Here, we investigated the mechanism of ARID4B function in mESC differentiation by focusing on genes and pathways that are upregulated in its absence. Material and Methods We analyzed transcriptomic results of wild-type and arid4bΔ endoderm or mesoderm differentiated cells through integrative genomics viewer and ingenuity pathway analysis. We performed real-time quantitative polymerase chain reaction for selected genes. To understand pathway activation, we performed Western blot for candidate proteins during the time-course of differentiation. We also analyzed H3K4me3, H3K27me3 and H3K27Ac ChIP-seq results to understand changes in the chromatin environment. Results Interferon-related genes were activated in arid4bΔ mESCs and endoderm or mesoderm directed cells. Consistent with this, higher phosphorylated STAT1 levels were found in arid4bΔ mESCs while a related phosphorylated STAT3 was unchanged. Finally, we observed a significant increase in H3K4me3 around interferon-related distal gene regulatory regions with a combination of either upregulation of H3K27Ac level or downregulation of H3K27me3 level. Conclusion These results provide evidence that ARID4B is involved in the suppression of interferon-related genes in mESCs and during meso/endoderm differentiation through modulation, mainly of H3K4me3. This regulation might be important for successful mESC differentiation.
Collapse
Affiliation(s)
- Nihal Terzi Çizmecioğlu
- Department of Biological Sciences, Middle East Technical University, Faculty of Arts and Sciences, Ankara, Turkey
| |
Collapse
|
6
|
Schneider P, Wander P, Arentsen-Peters STCJM, Vrenken KS, Rockx-Brouwer D, Adriaanse FRS, Hoeve V, Paassen I, Drost J, Pieters R, Stam RW. CRISPR-Cas9 Library Screening Identifies Novel Molecular Vulnerabilities in KMT2A-Rearranged Acute Lymphoblastic Leukemia. Int J Mol Sci 2023; 24:13207. [PMID: 37686014 PMCID: PMC10487613 DOI: 10.3390/ijms241713207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
In acute lymphoblastic leukemia (ALL), chromosomal translocations involving the KMT2A gene represent highly unfavorable prognostic factors and most commonly occur in patients less than 1 year of age. Rearrangements of the KMT2A gene drive epigenetic changes that lead to aberrant gene expression profiles that strongly favor leukemia development. Apart from this genetic lesion, the mutational landscape of KMT2A-rearranged ALL is remarkably silent, providing limited insights for the development of targeted therapy. Consequently, identifying potential therapeutic targets often relies on differential gene expression, yet the inhibition of these genes has rarely translated into successful therapeutic strategies. Therefore, we performed CRISPR-Cas9 knock-out screens to search for genetic dependencies in KMT2A-rearranged ALL. We utilized small-guide RNA libraries directed against the entire human epigenome and kinome in various KMT2A-rearranged ALL, as well as wild-type KMT2A ALL cell line models. This screening approach led to the discovery of the epigenetic regulators ARID4B and MBD3, as well as the receptor kinase BMPR2 as novel molecular vulnerabilities and attractive therapeutic targets in KMT2A-rearranged ALL.
Collapse
Affiliation(s)
- Pauline Schneider
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Priscilla Wander
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | - Kirsten S. Vrenken
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | | | - Veerle Hoeve
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Irene Paassen
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Ronald W. Stam
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
7
|
Das U, Gangisetty O, Chaudhary S, Tarale P, Rousseau B, Price J, Frazier I, Sarkar DK. Epigenetic insight into effects of prenatal alcohol exposure on stress axis development: Systematic review with meta-analytic approaches. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:18-35. [PMID: 36341762 DOI: 10.1111/acer.14972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
We conducted a systematic review with meta-analytic elements using publicly available Gene Expression Omnibus (GEO) datasets to determine the role of epigenetic mechanisms in prenatal alcohol exposure (PAE)-induced hypothalamic-pituitary-adrenal (HPA) axis dysfunctions in offspring. Several studies have demonstrated that PAE has long-term consequences on HPA axis functions in offspring. Some studies determined that alcohol-induced epigenetic alterations during fetal development persist in adulthood. However, additional research is needed to understand the major epigenetic events leading to alcohol-induced teratogenesis of the HPA axis. Our network analysis of GEO datasets identified key pathways relevant to alcohol-mediated histone modifications, DNA methylation, and miRNA involvement associated with PAE-induced alterations of the HPA axis. Our analysis indicated that PAE perturbated the epigenetic machinery to activate corticotrophin-releasing hormone, while it suppressed opioid, glucocorticoid receptor, and circadian clock genes. These results help to further our understanding of the epigenetic basis of alcohol's effects on HPA axis development.
Collapse
Affiliation(s)
- Ujjal Das
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Omkaram Gangisetty
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Shaista Chaudhary
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Prashant Tarale
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Bénédicte Rousseau
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Julianne Price
- Molecular Neuroscience of Alcohol and Drug Abuse Research Training, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Center of Alcohol & Substance Use Studies, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Department of Kinesiology & Health, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Ian Frazier
- Molecular Neuroscience of Alcohol and Drug Abuse Research Training, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Center of Alcohol & Substance Use Studies, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Department of Kinesiology & Health, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Dipak K Sarkar
- Endocrinology Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Molecular Neuroscience of Alcohol and Drug Abuse Research Training, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Center of Alcohol & Substance Use Studies, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Rutgers Endocrinology Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
8
|
Fidler G, Szilágyi-Rácz AA, Dávid P, Tolnai E, Rejtő L, Szász R, Póliska S, Biró S, Paholcsek M. Circulating microRNA sequencing revealed miRNome patterns in hematology and oncology patients aiding the prognosis of invasive aspergillosis. Sci Rep 2022; 12:7144. [PMID: 35504997 PMCID: PMC9065123 DOI: 10.1038/s41598-022-11239-z] [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: 09/09/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Invasive aspergillosis (IA) may occur as a serious complication of hematological malignancy. Delays in antifungal therapy can lead to an invasive disease resulting in high mortality. Currently, there are no well-established blood circulating microRNA biomarkers or laboratory tests which can be used to diagnose IA. Therefore, we aimed to define dysregulated miRNAs in hematology and oncology (HO) patients to identify biomarkers predisposing disease. We performed an in-depth analysis of high-throughput small transcriptome sequencing data obtained from the whole blood samples of our study cohort of 50 participants including 26 high-risk HO patients and 24 controls. By integrating in silico bioinformatic analyses of small noncoding RNA data, 57 miRNAs exhibiting significant expression differences (P < 0.05) were identified between IA-infected patients and non-IA HO patients. Among these, we found 36 differentially expressed miRNAs (DEMs) irrespective of HO malignancy. Of the top ranked DEMs, we found 14 significantly deregulated miRNAs, whose expression levels were successfully quantified by qRT-PCR. MiRNA target prediction revealed the involvement of IA related miRNAs in the biological pathways of tumorigenesis, the cell cycle, the immune response, cell differentiation and apoptosis.
Collapse
Affiliation(s)
- Gábor Fidler
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Anna Anita Szilágyi-Rácz
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Péter Dávid
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Emese Tolnai
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - László Rejtő
- Department of Hematology, Jósa András Teaching Hospital, Nyíregyháza, Hungary
| | - Róbert Szász
- Division of Hematology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Biró
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Melinda Paholcsek
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary.
| |
Collapse
|
9
|
Differentiation of fetal hematopoietic stem cells requires ARID4B to restrict autocrine KITLG/KIT-Src signaling. Cell Rep 2021; 37:110036. [PMID: 34818550 PMCID: PMC8722094 DOI: 10.1016/j.celrep.2021.110036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 07/15/2021] [Accepted: 11/01/2021] [Indexed: 11/26/2022] Open
Abstract
Balance between the hematopoietic stem cell (HSC) duality to either possess self-renewal capacity or differentiate into multipotency progenitors (MPPs) is crucial for maintaining homeostasis of the hematopoietic stem/progenitor cell (HSPC) compartment. To retain the HSC self-renewal activity, KIT, a receptor tyrosine kinase, in HSCs is activated by its cognate ligand KITLG originating from niche cells. Here, we show that AT-rich interaction domain 4B (ARID4B) interferes with KITLG/KIT signaling, consequently allowing HSC differentiation. Conditional Arid4b knockout in mouse hematopoietic cells blocks fetal HSC differentiation, preventing hematopoiesis. Mechanistically, ARID4B-deficient HSCs self-express KITLG and overexpress KIT. As to downstream pathways of KITLG/KIT signaling, inhibition of Src family kinases rescues the HSC differentiation defect elicited by ARID4B loss. In summary, the intrinsic ARID4B-KITLG/KIT-Src axis is an HSPC regulatory program that enables the differentiation state, while KIT stimulation by KITLG from niche cells preserves the HSPC undifferentiated pool. Hematopoietic stem cells (HSCs) at the top of the hematopoietic hierarchy are able to self-renew and differentiate to mature blood cells. Young et al. report that an HSC self-control mechanism established by ARID4B ensures HSC differentiation. ARID4B-deficient HSCs produce KITLG to stimulate KIT, leading to blockage of HSC differentiation and eventual hematopoietic failure.
Collapse
|
10
|
Keskin EG, Huang J, Terzi Çizmecioğlu N. Arid4b physically interacts with Tfap2c in mouse embryonic stem cells. Turk J Biol 2021; 45:162-170. [PMID: 33907498 PMCID: PMC8068770 DOI: 10.3906/biy-2010-67] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
Precise regulation of gene expression is required for embryonic stem cell (ESC) differentiation. Transcription factor (TF) networks coordinate the balance of pluripotency and differentiation in response to extracellular and intracellular signals. Chromatin factors work alongside TFs to achieve timely regulation of gene expression for differentiation process. Our previous studies showed that a member of the Sin3a corepressor complex, Arid4b, is critical for proper mouse ESC differentiation into mesoderm and endoderm. We found elevated histone 3 lysine 27 acetylation (H3K27Ac) in a subset of genomic loci in meso/endoderm directed arid4bΔ cells, coincident with their derepression. We reasoned that Sin3a complex may be required for the suppression of these genes during differentiation. To identify TFs that might cooperate with Arid4b for this function, we found consensus TF binding sequences enriched in H3K27Ac elevated regions in arid4bΔ cells. Of these candidate TFs, we validated expression of Bach1, Ddit3, Prrx2, Znf354c and Tfap2c in mESCs. We then demonstrated a physical interaction between Arid4b and Tfap2c in mESCs using endogenous coimmunoprecipitation and proximity ligation assay experiments. Our results point to a role of Arid4b in the Sin3a complex in repression of a subset of Tfap2c-regulated genes during meso/endoderm differentiation.
Collapse
Affiliation(s)
- Ezgi Gül Keskin
- Department of Biological Sciences, Faculty of Arts and Sciences, Middle East Technical University, Ankara Turkey
| | - Jialiang Huang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian China
| | - Nihal Terzi Çizmecioğlu
- Department of Biological Sciences, Faculty of Arts and Sciences, Middle East Technical University, Ankara Turkey
| |
Collapse
|
11
|
GÜVEN G, TERZİ ÇİZMECİOĞLU N. Arid4b alters cell cycle and cell death dynamics during mouse embryonic stem cell differentiation. Turk J Biol 2021; 45:56-64. [PMID: 33597822 PMCID: PMC7877710 DOI: 10.3906/biy-2009-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 01/23/2023] Open
Abstract
Cell division and death play an important role in embryonic development. Cell specialization is accompanied with slow proliferation and quiescence. Cell death is important for morphogenesis. Gene expression changes during differentiation is coordinated by lineage-specific transcription factors and chromatin factors. It is not yet fully understood how alterations in gene expression and cell cycle/death mechanisms are connected. We previously identified a chromatin protein Arid4b as a critical factor for meso/endoderm differentiation of mouse embryonic stem cells (mESCs). The differentiation defect of Arid4b-deficient mESCs might be due to misregulation of cell proliferation or death. Here, we identified a role for Arid4b in cell cycle rewiring at the onset of differentiation. Arid4b-deficient differentiating cells have less proliferative capacity and their cell cycle profile is more similar to mESC stage than the differentiating wild-type cells. We found no evidence of increased DNA damage or checkpoint activation. Our investigation of cell death mechanisms found no contribution from autophagy but revealed a slight increase in Caspase-3 activation implying early apoptosis in Arid4b-deficient differentiating cells. Taken together, our data suggest Arid4b regulates cell cycle alterations during exit from pluripotency. Future studies will be instrumental in understanding whether these changes directly contribute to Arid4b-dependent differentiation control.
Collapse
Affiliation(s)
- Gözde GÜVEN
- Department of Biological Sciences, Faculty of Arts and Sciences, Middle East Technical University, AnkaraTurkey
| | - Nihal TERZİ ÇİZMECİOĞLU
- Department of Biological Sciences, Faculty of Arts and Sciences, Middle East Technical University, AnkaraTurkey
| |
Collapse
|