Loss of the Transcription Factor MAFB Limits β-Cell

Total Page:16

File Type:pdf, Size:1020Kb

Loss of the Transcription Factor MAFB Limits β-Cell UCSF UC San Francisco Previously Published Works Title Loss of the transcription factor MAFB limits β-cell derivation from human PSCs. Permalink https://escholarship.org/uc/item/0zb338n9 Journal Nature communications, 11(1) ISSN 2041-1723 Authors Russell, Ronan Carnese, Phichitpol P Hennings, Thomas G et al. Publication Date 2020-06-02 DOI 10.1038/s41467-020-16550-9 Peer reviewed eScholarship.org Powered by the California Digital Library University of California ARTICLE https://doi.org/10.1038/s41467-020-16550-9 OPEN Loss of the transcription factor MAFB limits β-cell derivation from human PSCs Ronan Russell1, Phichitpol P. Carnese1, Thomas G. Hennings1, Emily M. Walker 2, Holger A. Russ 1,3, ✉ Jennifer S. Liu1, Simone Giacometti1, Roland Stein2 & Matthias Hebrok 1 Next generation sequencing studies have highlighted discrepancies in β-cells which exist between mice and men. Numerous reports have identified MAF BZIP Transcription Factor B 1234567890():,; (MAFB) to be present in human β-cells postnatally, while its expression is restricted to embryonic and neo-natal β-cells in mice. Using CRISPR/Cas9-mediated gene editing, coupled with endocrine cell differentiation strategies, we dissect the contribution of MAFB to β-cell development and function specifically in humans. Here we report that MAFB knockout hPSCs have normal pancreatic differentiation capacity up to the progenitor stage, but favor soma- tostatin- and pancreatic polypeptide–positive cells at the expense of insulin- and glucagon- producing cells during endocrine cell development. Our results describe a requirement for MAFB late in the human pancreatic developmental program and identify it as a distinguishing transcription factor within islet cell subtype specification. We propose that hPSCs represent a powerful tool to model human pancreatic endocrine development and associated disease pathophysiology. 1 UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA. 2 Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA. 3Present address: Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Denver, ✉ Aurora, CO 80045, USA. email: [email protected] NATURE COMMUNICATIONS | (2020) 11:2742 | https://doi.org/10.1038/s41467-020-16550-9 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16550-9 dentifying key factors that mediate islet cell function is critical In the present study, we generate isogenic MAFB loss of Ito understanding Type 1 and Type 2 Diabetes Mellitus (T1D, function hPSCs to dissect its contribution in human β-cell T2D) disease processes and to moving forward with cell-based development, using a stepwise differentiation protocol. MAFB interventions in humans1. Notably, the advent of in-depth loss of function cells display no major defects in the ability to sequencing studies has elucidated alternative transcription fac- specify early stages of pancreatic development. However, MAFB tor activity among closely related islet cell subtypes2,3 and addi- null cells have an increased propensity to generate somatostatin- tionally, uncovered numerous examples of heterogeneity within (δ) and pancreatic polypeptide- (γ) producing cells at the expense healthy β-cells4. It has long been recognized that differences in of glucagon (α) and insulin (β) producing cells. Our results cellular composition and architecture exist between human and identify an important role of MAFB within endocrine cell- mice adult islets, although the precise reasons and implications subtype specification decisions and demonstrate that it is a critical for such disparity have remained elusive. Numerous recent single- transcription factor for the generation of glucose-responsive β- cell RNA-sequencing studies have reported a discrepancy in the cells from hPSCs. This essential role of MAFB in human β-cell expression of a number of factors, highlighting potential species- identity is considerably different from what was concluded in specific regulatory differences among rodent and human β-cells/ genetically engineered mouse models (GEMMs). These findings islet counterparts3,5. suggest that species-specific differences in transcription factor One such factor is MAFB, which has been identified in activity may exist between mice and men and promote the use of numerous studies to be expressed within adult human β-cells, human developmental models such as directed differentiation while being absent from the murine counterparts. MAFB is a strategies to enhance our understanding of human biology. member of the large Maf family of transcription factors that share highly similar basic region/leucine zipper DNA binding motifs Results and N-terminal activation domains. MAFB is expressed in MAFB is expressed during β-like cell differentiation.We developing pancreatic α- and β-cells in the mouse, while its – employed our previously published β-cell directed differentiation expression becomes restricted to α-cells in the adult6 8 and protocol with modifications (Fig. 1a, Supplementary Tables 1 and MAFB knockout (KO) in the mouse pancreas merely delays β-cell 2)23,24. Utilizing the INS-GFP reporter hESC line, in which GFP maturation for a short period of time9,10. MAFB binds to the is knocked into one endogenous locus of the insulin gene25, this MAF responsive element (MARE) expressed within critical β-cell protocol routinely generates >90% definitive endoderm (DE) cells genes including PDX1 and INS. Kim and colleagues recently co-expressing SOX17 and FOXA2, >70% PP cells co-expressing showed that MAFB mRNA is expressed at robust levels both in α- PDX1 and NKX6.1, and 20–50% mono-hormonal β-like cells and β-cells in humans with no significant fluctuations in levels (β-like) co-expressing C-peptide and NKX6.1 as assessed by flow over age11,afinding that has since been demonstrated at protein cytometry (FC) and immunofluorescence (IF) staining (Fig. 1b, c, level12. Building on this, single-cell RNA-sequencing studies have Supplementary Fig. 1a). First, we examined the expression of identified that MAFB is robustly expressed in human adult β-cells – MAFB over the time course of differentiation using qPCR and as well as α- and δ-cells2,3,5,13 16. This is a notable difference to western blot analysis. mRNA levels of MAFB are first detected at the related factor MAFA, a β-cell specific transcription factor that the PP stage and increase in levels at the β-like stage (Fig. 1d). also binds to the insulin MARE site, activates insulin gene tran- MAFB protein was principally detected at the β-like stage, indi- scription7, and has been reported as a critical factor in adult β- cating that robust MAFB expression coincides with the onset of cells across species. MAFA expression increases in an age- hormone expression (Fig. 1e)7. dependent manner in both human and mouse β-cells and is the In order to examine the role of MAFB in the differentiation of predominant isoform expressed in functional mouse β-cells12, β-cells, we utilized an inducible (i)CRISPR gene editing system26 although it is not robustly expressed in human β-cells until after in the INS-GFP cell line along with three individual guide RNAs ~10 years of age3,11. Together, these data suggest a potential (gRNAs) (Fig. 1f, Supplementary Table 3). Three independent evolutionary discrepancy in the role of MAFB in the pancreatic MAFB heterozygous and homozygous clones with frameshift islet compartment and the use of MAFA as a marker of human β- mutations in the transactivation domain, leading to disruption of cell function has been questioned in recent studies17. the functional full-length protein, were established. Isogenic Directed differentiation of human pluripotent stem cells clonal cell lines which lacked genomic editing were employed as (hPSCs) into insulin producing β-like cells provides a powerful controls. All clonal hESC lines grew as expected compared with platform to investigate human biology. This approach has been the parental control line, suggesting the genomic editing and sub- shown to effectively recapitulate the sequential stages of islet cell cloning procedure did not compromise pluripotency. To verify development, in which hPSCs transition through a definitive the genotypes, cell lines were assayed via targeted sequencing of endoderm (DE) stage, multipotent pancreatic progenitor (PP) the genome-edited locus at the pluripotency stage, as well as by stage, and toward β-like cells. By combining this strategy with western blot and IF analysis for MAFB protein at the β-like cell genome editing approaches such as CRISPR/Cas9 (clustered stage (Fig. 1g–h, Supplementary Fig. 1b). Mono-allelic and bi- regularly interspaced short palindromic repeats), it is possible to allelic MAFB clonal lines harboring frameshift mutations will be dissect the specific contribution of pancreatic transcription fac- referred to as +/− and −/−, respectively, throughout the text and tors at precise stages in a human-specific context18. While much specific indel sequence information for each clone is outlined in effort has been spent focusing on optimizing directed differ- Supplementary Tables 4 and 5. entiation of human cell types for disease modeling or the phe- notypes associated with known genetic susceptibility genes19,20, the underlying complex biological processes which established Differentiation of MAFB KO hESCs to pancreatic β-like cells. protocols recapitulate also provides a unique platform for To control for potential off-target
Recommended publications
  • Activated Peripheral-Blood-Derived Mononuclear Cells
    Transcription factor expression in lipopolysaccharide- activated peripheral-blood-derived mononuclear cells Jared C. Roach*†, Kelly D. Smith*‡, Katie L. Strobe*, Stephanie M. Nissen*, Christian D. Haudenschild§, Daixing Zhou§, Thomas J. Vasicek¶, G. A. Heldʈ, Gustavo A. Stolovitzkyʈ, Leroy E. Hood*†, and Alan Aderem* *Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103; ‡Department of Pathology, University of Washington, Seattle, WA 98195; §Illumina, 25861 Industrial Boulevard, Hayward, CA 94545; ¶Medtronic, 710 Medtronic Parkway, Minneapolis, MN 55432; and ʈIBM Computational Biology Center, P.O. Box 218, Yorktown Heights, NY 10598 Contributed by Leroy E. Hood, August 21, 2007 (sent for review January 7, 2007) Transcription factors play a key role in integrating and modulating system. In this model system, we activated peripheral-blood-derived biological information. In this study, we comprehensively measured mononuclear cells, which can be loosely termed ‘‘macrophages,’’ the changing abundances of mRNAs over a time course of activation with lipopolysaccharide (LPS). We focused on the precise mea- of human peripheral-blood-derived mononuclear cells (‘‘macro- surement of mRNA concentrations. There is currently no high- phages’’) with lipopolysaccharide. Global and dynamic analysis of throughput technology that can precisely and sensitively measure all transcription factors in response to a physiological stimulus has yet to mRNAs in a system, although such technologies are likely to be be achieved in a human system, and our efforts significantly available in the near future. To demonstrate the potential utility of advanced this goal. We used multiple global high-throughput tech- such technologies, and to motivate their development and encour- nologies for measuring mRNA levels, including massively parallel age their use, we produced data from a combination of two distinct signature sequencing and GeneChip microarrays.
    [Show full text]
  • FOXA2 Is a Sensitive and Specific Marker for Small Cell Neuroendocrine Carcinoma of the Prostate Jung Wook Park1, John K Lee2,3, Owen N Witte1,4,5 and Jiaoti Huang6
    Modern Pathology (2017) 30, 1262–1272 1262 © 2017 USCAP, Inc All rights reserved 0893-3952/17 $32.00 FOXA2 is a sensitive and specific marker for small cell neuroendocrine carcinoma of the prostate Jung Wook Park1, John K Lee2,3, Owen N Witte1,4,5 and Jiaoti Huang6 1Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA; 2Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA; 3Molecular Biology Institute, David Geffen School of Medicine, University of California— Los Angeles, Los Angeles, CA, USA; 4Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California—Los Angeles, Los Angeles, CA, USA; 5Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA and 6Department of Pathology, Duke University School of Medicine, Durham, NC, USA The median survival of patients with small cell neuroendocrine carcinoma is significantly shorter than that of patients with classic acinar-type adenocarcinoma. Small cell neuroendocrine carcinoma is traditionally diagnosed based on histologic features because expression of current immunohistochemical markers is inconsistent. This is a challenging diagnosis even for expert pathologists and particularly so for pathologists who do not specialize in prostate cancer. New biomarkers to aid in the diagnosis of small cell neuroendocrine carcinoma are therefore urgently needed. We discovered that FOXA2, a pioneer transcription factor, is frequently and specifically expressed in small cell neuroendocrine carcinoma compared with prostate adenocarcinoma from published mRNA-sequencing data of a wide range of human prostate cancers.
    [Show full text]
  • Charting Brachyury-Mediated Developmental Pathways During Early Mouse Embryogenesis
    Charting Brachyury-mediated developmental pathways during early mouse embryogenesis Macarena Lolasa,b, Pablo D. T. Valenzuelab, Robert Tjiana,c,1, and Zhe Liud,1 dJunior Fellow Program, aJanelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147; bFundación Ciencia para la Vida, Santiago 7780272, Chile; and cLi Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine Center of Excellence, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720 Contributed by Robert Tjian, February 11, 2014 (sent for review January 14, 2014) To gain insights into coordinated lineage-specification and mor- cells play diverse and indispensable roles in early mouse phogenetic processes during early embryogenesis, here we report development. a systematic identification of transcriptional programs mediated In mouse ES cell-based differentiation systems, Brachyury is by a key developmental regulator—Brachyury. High-resolution widely used as a mesoendoderm marker, and Brachyury-positive chromosomal localization mapping of Brachyury by ChIP sequenc- cells were able to differentiate into mesodermal and definitive ing and ChIP-exonuclease revealed distinct sequence signatures endodermal lineages, such as cardiomyocytes and hepatocytes enriched in Brachyury-bound enhancers. A combination of genome- (8–10). In a previous study, we found that depletion of a core wide in vitro and in vivo perturbation analysis and cross-species promoter factor, the TATA binding protein-associated factor 3, evolutionary comparison unveiled a detailed Brachyury-depen- in ES cells leads to significant up-regulation of Brachyury and dent gene-regulatory network that directly links the function of mesoderm lineages during ES cell differentiation (11). Here, we Brachyury to diverse developmental pathways and cellular house- systematically characterized the molecular function of Brachyury keeping programs.
    [Show full text]
  • Prospective Isolation of NKX2-1–Expressing Human Lung Progenitors Derived from Pluripotent Stem Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE Prospective isolation of NKX2-1–expressing human lung progenitors derived from pluripotent stem cells Finn Hawkins,1,2 Philipp Kramer,3 Anjali Jacob,1,2 Ian Driver,4 Dylan C. Thomas,1 Katherine B. McCauley,1,2 Nicholas Skvir,1 Ana M. Crane,3 Anita A. Kurmann,1,5 Anthony N. Hollenberg,5 Sinead Nguyen,1 Brandon G. Wong,6 Ahmad S. Khalil,6,7 Sarah X.L. Huang,3,8 Susan Guttentag,9 Jason R. Rock,4 John M. Shannon,10 Brian R. Davis,3 and Darrell N. Kotton1,2 2 1Center for Regenerative Medicine, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA. 3Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA. 4Department of Anatomy, UCSF, San Francisco, California, USA. 5Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA. 6Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA. 7Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA. 8Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA. 9Department of Pediatrics, Monroe Carell Jr. Children’s Hospital, Vanderbilt University, Nashville, Tennessee, USA. 10Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA. It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1–expressing (NKX2-1+) precursor cells.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • Utx Is Required for Proper Induction of Ectoderm and Mesoderm During Differentiation of Embryonic Stem Cells
    Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells Cristina Morales Torres1,2, Anne Laugesen1,2, Kristian Helin1,2,3* 1 Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark, 2 Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark, 3 The Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark Abstract Embryonic development requires chromatin remodeling for dynamic regulation of gene expression patterns to ensure silencing of pluripotent transcription factors and activation of developmental regulators. Demethylation of H3K27me3 by the histone demethylases Utx and Jmjd3 is important for the activation of lineage choice genes in response to developmental signals. To further understand the function of Utx in pluripotency and differentiation we generated Utx knockout embryonic stem cells (ESCs). Here we show that Utx is not required for the proliferation of ESCs, however, Utx contributes to the establishment of ectoderm and mesoderm in vitro. Interestingly, this contribution is independent of the catalytic activity of Utx. Furthermore, we provide data showing that the Utx homologue, Uty, which is devoid of detectable demethylase activity, and Jmjd3 partly compensate for the loss of Utx. Taken together our results show that Utx is required for proper formation of ectoderm and mesoderm in vitro, and that Utx, similar to its C.elegans homologue, has demethylase dependent and independent functions. Citation: Morales Torres C, Laugesen A, Helin K (2013) Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells. PLoS ONE 8(4): e60020. doi:10.1371/journal.pone.0060020 Editor: Qiang Wu, National University of Singapore, Singapore Received December 13, 2012; Accepted February 21, 2013; Published April 3, 2013 Copyright: ß 2013 Morales Torres et al.
    [Show full text]
  • Thyroid Gland Development and Function in the Zebrafish Model P
    THYROID GLAND DEVELOPMENT AND FUNCTION IN THE ZEBRAFISH MODEL P. Porazzi, D. Calebiro, F. Benato, N. Tiso, L. Persani To cite this version: P. Porazzi, D. Calebiro, F. Benato, N. Tiso, L. Persani. THYROID GLAND DEVELOPMENT AND FUNCTION IN THE ZEBRAFISH MODEL. Molecular and Cellular Endocrinology, Elsevier, 2009, 312 (1-2), pp.14. 10.1016/j.mce.2009.05.011. hal-00521550 HAL Id: hal-00521550 https://hal.archives-ouvertes.fr/hal-00521550 Submitted on 28 Sep 2010 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Accepted Manuscript Title: THYROID GLAND DEVELOPMENT AND FUNCTION IN THE ZEBRAFISH MODEL Authors: P. Porazzi, D. Calebiro, F. Benato, N. Tiso, L. Persani PII: S0303-7207(09)00277-9 DOI: doi:10.1016/j.mce.2009.05.011 Reference: MCE 7224 To appear in: Molecular and Cellular Endocrinology Received date: 6-3-2009 Revised date: 20-5-2009 Accepted date: 20-5-2009 Please cite this article as: Porazzi, P., Calebiro, D., Benato, F., Tiso, N., Persani, L., THYROID GLAND DEVELOPMENT AND FUNCTION IN THE ZEBRAFISH MODEL, Molecular and Cellular Endocrinology (2008), doi:10.1016/j.mce.2009.05.011 This is a PDF file of an unedited manuscript that has been accepted for publication.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Zfp57 Mutant ES Cell Lines Directly Derived from Blastocysts
    Stem Cell Research 16 (2016) 282–286 Contents lists available at ScienceDirect Stem Cell Research journal homepage: www.elsevier.com/locate/scr Lab Resource: Stem Cell Line Zfp57 mutant ES cell lines directly derived from blastocysts Ho-Tak Lau a, Lizhi Liu a, Xiajun Li a,b,⁎ a Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA b Department of Oncological Sciences, Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA article info abstract Article history: Zfp57 is a master regulator of genomic imprinting in mouse embryos. To further test its functions, we have Received 25 November 2015 derived multiple Zfp57 mutant ES clones directly from mouse blastocysts. Indeed, we found DNA methylation im- Received in revised form 30 December 2015 print was lost at most examined imprinting control regions in these Zfp57 mutant ES clones, similar to what was Accepted 31 December 2015 observed in Zfp57 mutant embryos in the previous studies. This result indicates that these blastocyst-derived Available online 6 January 2016 Zfp57 mutant ES clones can be employed for functional analyses of Zfp57 in genomic imprinting. © 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Resource table heterozygous female mice and Zfp57−/− homozygous male mice, whereas five Zfp57−/− (M−Z−) ES clones were derived from the cross between Zfp57−/− homozygous female mice and Zfp57−/− homozygous male mice.
    [Show full text]
  • Estrogen Receptor Α AF-2 Mutation Results in Antagonist Reversal and Reveals Tissue Selective Function of Estrogen Receptor Modulators
    Estrogen receptor α AF-2 mutation results in antagonist reversal and reveals tissue selective function of estrogen receptor modulators Yukitomo Araoa, Katherine J. Hamiltona, Manas K. Rayb, Gregory Scottb, Yuji Mishinac, and Kenneth S. Koracha,1 aReceptor Biology Section, Laboratory of Reproductive and Developmental Toxicology and bKnock Out Core, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC 27709; and cSchool of Dentistry, University of Michigan, Ann Arbor, MI 48109 Edited by David J. Mangelsdorf, University of Texas Southwestern Medical Center, Dallas, TX, and approved July 22, 2011 (received for review June 10, 2011) The estrogen receptor (ER) is a ligand-dependent transcription and that may be related to the cell type specific functionality factor containing two transcriptional activation domains. AF-1 is in of TAM (12). However, it is still not entirely clear how TAM the N terminus of the receptor protein and AF-2 activity is manifests agonist activities through ERα WT in different tissues. dependent on helix 12 of the C-terminal ligand-binding domain. We focused on the L543A and L544A mutations in the ERα Two point mutations of leucines 543 and 544 to alanines (L543A, AF-2 domain (AF2ER) to evaluate the ERα AF-1 and AF-2 L544A) in helix 12 minimized estrogen-dependent transcriptional functions in vivo and the SERM functionality in the tissues. α α activation and reversed the activity of the estrogen antagonists The ER -KO ( ERKO) mouse is an established model for α α ICI182780 (ICI) and tamoxifen (TAM) into agonists in a similar evaluating ER function in vivo.
    [Show full text]
  • Potential Genotoxicity from Integration Sites in CLAD Dogs Treated Successfully with Gammaretroviral Vector-Mediated Gene Therapy
    Gene Therapy (2008) 15, 1067–1071 & 2008 Nature Publishing Group All rights reserved 0969-7128/08 $30.00 www.nature.com/gt SHORT COMMUNICATION Potential genotoxicity from integration sites in CLAD dogs treated successfully with gammaretroviral vector-mediated gene therapy M Hai1,3, RL Adler1,3, TR Bauer Jr1,3, LM Tuschong1, Y-C Gu1,XWu2 and DD Hickstein1 1Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA and 2Laboratory of Molecular Technology, Scientific Applications International Corporation-Frederick, National Cancer Institute-Frederick, Frederick, Maryland, USA Integration site analysis was performed on six dogs with in hematopoietic stem cells. Integrations clustered around canine leukocyte adhesion deficiency (CLAD) that survived common insertion sites more frequently than random. greater than 1 year after infusion of autologous CD34+ bone Despite potential genotoxicity from RIS, to date there has marrow cells transduced with a gammaretroviral vector been no progression to oligoclonal hematopoiesis and no expressing canine CD18. A total of 387 retroviral insertion evidence that vector integration sites influenced cell survival sites (RIS) were identified in the peripheral blood leukocytes or proliferation. Continued follow-up in disease-specific from the six dogs at 1 year postinfusion. A total of 129 RIS animal models such as CLAD will be required to provide an were identified in CD3+ T-lymphocytes and 102 RIS in accurate estimate
    [Show full text]
  • Mechanism of Nucleosome Targeting by Pioneer Transcription Factors
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2019 Mechanism Of Nucleosome Targeting By Pioneer Transcription Factors Meilin Mary Fernandez Garcia University of Pennsylvania Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Biochemistry Commons, and the Biophysics Commons Recommended Citation Fernandez Garcia, Meilin Mary, "Mechanism Of Nucleosome Targeting By Pioneer Transcription Factors" (2019). Publicly Accessible Penn Dissertations. 3624. https://repository.upenn.edu/edissertations/3624 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/3624 For more information, please contact [email protected]. Mechanism Of Nucleosome Targeting By Pioneer Transcription Factors Abstract Transcription factors (TFs) forage the genome to instruct cell plasticity, identity, and differentiation. These developmental processes are elicited through TF engagement with chromatin. Yet, how and which TFs can engage with chromatin and thus, nucleosomes, remains largely unexplored. Pioneer TFs are TF that display a high affinity for nucleosomes. Extensive genetic and biochemical studies on the pioneer TF FOXA, a driver of fibroblast to hepatocyte reprogramming, revealed its nucleosome binding ability and chromatin targeting lead to chromatin accessibility and subsequent cooperative binding of TFs. Similarly, a number of reprogramming TFs have been suggested to have pioneering activity due to their ability to target compact chromatin and increase accessibility and enhancer formation in vivo. But whether these factors directly interact with nucleosomes remains to be assessed. Here we test the nucleosome binding ability of the cell reprogramming TFs, Oct4, Sox2, Klf4 and cMyc, that are required for the generation of induced pluripotent stem cells. In addition, we also test neuronal and macrophage reprogramming TFs.
    [Show full text]