Single Cell RNA Sequencing Identifies Early Diversity of Sensory Neurons

Total Page:16

File Type:pdf, Size:1020Kb

Single Cell RNA Sequencing Identifies Early Diversity of Sensory Neurons ARTICLE https://doi.org/10.1038/s41467-020-17929-4 OPEN Single cell RNA sequencing identifies early diversity of sensory neurons forming via bi-potential intermediates Louis Faure 1, Yiqiao Wang2, Maria Eleni Kastriti 1, Paula Fontanet 2, Kylie K. Y. Cheung2, Charles Petitpré2, Haohao Wu2, Lynn Linyu Sun2, Karen Runge3, Laura Croci 4, Mark A. Landy 5, Helen C. Lai 5, Gian Giacomo Consalez4, Antoine de Chevigny 3, François Lallemend2,6, ✉ Igor Adameyko 1,7 & Saida Hadjab 2 1234567890():,; Somatic sensation is defined by the existence of a diversity of primary sensory neurons with unique biological features and response profiles to external and internal stimuli. However, there is no coherent picture about how this diversity of cell states is transcriptionally gen- erated. Here, we use deep single cell analysis to resolve fate splits and molecular biasing processes during sensory neurogenesis in mice. Our results identify a complex series of successive and specific transcriptional changes in post-mitotic neurons that delineate hier- archical regulatory states leading to the generation of the main sensory neuron classes. In addition, our analysis identifies previously undetected early gene modules expressed long before fate determination although being clearly associated with defined sensory subtypes. Overall, the early diversity of sensory neurons is generated through successive bi-potential intermediates in which synchronization of relevant gene modules and concurrent repression of competing fate programs precede cell fate stabilization and final commitment. 1 Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria. 2 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden. 3 INMED INSERM U1249, Aix-Marseille University, Marseille, France. 4 Università Vita-Salute San Raffaele, 20132 Milan, Italy. 5 Department of Neuroscience, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA. 6 Ming-Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden. 7 Department of Physiology and Pharmacology, Karolinska Institutet, ✉ 17177 Stockholm, Sweden. email: [email protected] NATURE COMMUNICATIONS | (2020) 11:4175 | https://doi.org/10.1038/s41467-020-17929-4 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17929-4 n the developing nervous system, various neuronal and glial the transcriptome of individual single cells (progenitors and cell types are generated from multipotent progenitor cells in a neurons) of the somatosensory neuro-glial progeny of the trunk. I fi + speci c chronological sequence. The continuous change in the For this, we FACS isolated tdTomato positive (TOM ) cells cellular potential of the progenitors is a key aspect to this isolated from mouse lines which selectively represent all NCCs developmental process1. As neuronal progenitors exit cell cycle, (Wnt1Cre;R26tdTOM and Plp1CreERT2;R26tdTOM) and from they differentiate into distinct neuron types that already provide neuron-specific Cre mouse lines (Isl1Cre;R26tdTOM and Ntrk3Cre; the underpinning for the identity of neuronal circuit in which R26tdTOM) to obtain an enrichment of somatosensory neuronal they will operate1,2. Hence, the formation of specific fates criti- populations (Supplementary Fig. 1a, b) from E9.5, E10.5, E11.5 cally depends on the timing and diversity of expression of and E12.5 and sequenced mRNAs of single cells with high cov- molecular factors in discrete parts of the nervous system3,4. erage using Smart-seq2 protocol (median of 8070 genes detected However, following this process, how the expression of molecular per cell) (Fig. 1a and Supplementary Fig. 1c, d; see “Methods”). determinants is progressively acquired and restricted to particular The selected stages correspond to key events of sensory lineage neurons as they further diversify into distinct neuronal classes development starting from migrating NCCs (E9.5) and finishing remains unclear. A holistic understanding of the stepwise with the end of the second wave of sensory neurogenesis and the execution of elaborate transcriptional programs for neuronal early specification of neuronal subtypes (E12.5). Our dataset diversification in heterogeneous system is still elusive. It is contains cells from Isl1Cre;R26tdTOM at E9.5 and E10.5 and therefore essential to dissect and understand the precise temporal Ntrk3Cre;R26tdTOM at E11.5 and E12.5, both lines label sensory progression of the gene-regulatory networks that produce and neurons, Wnt1Cre;R26tdTOM was used to label NCCs, glial and assemble neuronal complexity. Indeed, such complexity is found neuronal progenitors at E9.5 and E10.5 and Plp1CreERT2; in all parts of the developing nervous system, including the highly R26tdTOM at E12.5. The combination of cells collected after tra- heterogeneous population of sensory neurons—cells that live in cing with these Cre lines should provide a complete compendium dorsal root ganglia (DRG) and provide us with sensations of of neuronal progenitors and early neuronal subtypes in mouse touch, pain, itch, temperature and position in space5. DRG (for FACS gating strategies, see Supplementary Fig. 2). Co- The diversity of sensory modalities emerges during early embedding of the cells from all tracing strategies was made embryonic development as subtypes of sensory neurons are without tracing-based batch correction and lead to overlapping of generated from a common progenitor population, the neural crest cells of different tracings following both a developmental time- cells (NCCs). Two main waves of sensory neurogenesis in DRG wise trajectory and a progression from progenitors to specified occur between E9.5 and E13. The first wave of neurogenesis neurons (Fig. 1a–e and Supplementary Fig. 1a, b; see below for (E9.5−10.5)6 gives rise to myelinated neurons, the cutaneous low- trajectories) which together convincingly validated the use of the threshold mechanoreceptors (here after named mechan- dataset for further computational analysis. oreceptors) and the muscle proprioceptive afferents (here after We used RNA velocity10, an unbiased approach that leverages named proprioceptors) and a small population of Aδ-fibers that the distinction of spliced and unspliced RNA transcripts from the can be distinguished by E11.5 based on their specific expression aligned sequences, allowing us to obtain an additional timepoint of transmembrane receptors and transcription factors6,7. for each cells (t: expression of old spliced RNA and t + 1: Mechanoreceptors express RET and MAFA and/or TRKB, pro- expression of new unspliced RNA). Using the proportion between prioceptors express tropomyosin receptor kinase C (TRKC) and these two values for a given gene, and under some assumptions, it RUNX3, and the small Aδ-nociceptor population expresses is possible to infer whether the expression of a gene is being TRKA. Later, these populations will diversify even further, giving initiated or downregulated. By combining this information for all rise to additional subtypes representing the medium-to-large genes in each cell, as well as comparing it to its neighbor cells, it is diameter DRG neurons responsible for muscle proprioceptive possible to infer a direction vector indicating the putative future feedback and skin mechanosensation modalities5. A second wave transcriptional state of the cell. In our dataset, RNA velocity of neurogenesis (that peaks at E12 in a mouse embryo) generates revealed a clear differentiation directionality from NCCs to the small diameter unmyelinated nociceptive neurons5–7, which neuronal progenies. In order to recapitulate such transitions, the co-express TRKA and RUNX1 and later will diversify into sub- dataset was first projected onto diffusion space to denoise the types necessary for pain, itch and thermal sensation8 and inner- underlying geometry, and a principal tree has been fitted using vate the skin and deep tissues. Although the heterogeneity of ElPiGraph11 in a semi-supervised way with the help of clustering DRG neurons in adult mice are increasingly characterized8,9, the results (Fig. 1b, c). ElPiGraph is a manifold learning method logic and molecular mechanisms that allow progenitors and sen- which aims at inferring a principal graph (such as a tree) “passing sory neurons to fate split and unfold specific sensory neuronal through the middle of data” in high dimension. Cells were subprograms from sensory-biased NCCs remain to be ordered along the principal tree, and for each cell the distance on understood. the graph to a chosen root is considered as a pseudotime value. Here we use deep single-cell RNA sequencing (scRNAseq) Pseudotime analysis of gene expression showed significant and together with other experimental approaches to investigate fate robust changes along the reconstructed tree (8432 genes at a false choices, lineage relationships and molecular determinants during discovery rate (FDR) of 0.05), leading to the identification of early stages of sensory neurogenesis in the mouse. Overall, our major clusters (Source Data file). It also captured tree root data provide insights into the structure of the Waddingtonian populations and endpoints as well as intermediate states as sub- landscape of the sensory lineage and suggest that counteracting clusters, leading to the identification of two main neurogenic gene-regulatory networks operate within immature postmitotic trajectories named branch A and branch B (Fig. 1c). Those
Recommended publications
  • Combined Analysis of Chip-Seq and Gene Microarray Datasets Identify the E2-Mediated Genes in Erα-Dependent Manner in Osteosarcoma
    ONCOLOGY REPORTS 38: 2335-2342, 2017 Combined analysis of ChIP-seq and gene microarray datasets identify the E2-mediated genes in ERα-dependent manner in osteosarcoma KANGSONG TIAN1, WEI QI1, QIAN YAN1, FEnG ZHanG1, DELEI SONG1, HaIyanG ZHanG2 and MING LV1 1Trauma Department of Orthopedics, 2Microscopic Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China Received March 14, 2017; Accepted August 11, 2017 DOI: 10.3892/or.2017.5914 Abstract. Osteosarcoma is a common bone tumor which is Introduction affected by E2, the most representative estrogen. Gene regula- tion function of E2 is highly dependent on estrogen receptor. ESR1, also known as ER-alpha or ERα, is an important The purpose of this study was to explore the gene regulation estrogen receptor (ER) and involves in the gene regula- patterns of E2 through estrogen receptor α (ESR1) in osteo- tion in various diseases and biological processes, including sarcoma based on the combined analysis of ChIP-seq and breast cancer (1,2), osteosarcoma (3) and cell growth (4). gene microarray. All of the datasets were downloaded from 17β-estradiol (E2) is one of the most representative estrogens the Gene Expression Omnibus (GEO). Differential expression responsible for the development and reproductive capability genes (DEGs) in E2 treated U2OS cells expressing ESR1 of female characteristics (5). Besides, it participates in the (U2OS-ERα) compared with those treated with vehicle were progression of many diseases, for example, E2 could regulate obtained based on R programming software. ESR1-specific the proliferation of breast cancer cells through focal adhesion binding sites (peaks) in E2 treated U2OS cells were identified and chemokine signaling pathways (6); through autophagy through MACS.
    [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]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Viewed in [2, 3])
    Yildiz et al. Neural Development (2019) 14:5 https://doi.org/10.1186/s13064-019-0129-x RESEARCH ARTICLE Open Access Zebrafish prdm12b acts independently of nkx6.1 repression to promote eng1b expression in the neural tube p1 domain Ozge Yildiz1, Gerald B. Downes2 and Charles G. Sagerström1* Abstract Background: Functioning of the adult nervous system depends on the establishment of neural circuits during embryogenesis. In vertebrates, neurons that make up motor circuits form in distinct domains along the dorsoventral axis of the neural tube. Each domain is characterized by a unique combination of transcription factors (TFs) that promote a specific fate, while repressing fates of adjacent domains. The prdm12 TF is required for the expression of eng1b and the generation of V1 interneurons in the p1 domain, but the details of its function remain unclear. Methods: We used CRISPR/Cas9 to generate the first germline mutants for prdm12 and employed this resource, together with classical luciferase reporter assays and co-immunoprecipitation experiments, to study prdm12b function in zebrafish. We also generated germline mutants for bhlhe22 and nkx6.1 to examine how these TFs act with prdm12b to control p1 formation. Results: We find that prdm12b mutants lack eng1b expression in the p1 domain and also possess an abnormal touch-evoked escape response. Using luciferase reporter assays, we demonstrate that Prdm12b acts as a transcriptional repressor. We also show that the Bhlhe22 TF binds via the Prdm12b zinc finger domain to form a complex. However, bhlhe22 mutants display normal eng1b expression in the p1 domain. While prdm12 has been proposed to promote p1 fates by repressing expression of the nkx6.1 TF, we do not observe an expansion of the nkx6.1 domain upon loss of prdm12b function, nor is eng1b expression restored upon simultaneous loss of prdm12b and nkx6.1.
    [Show full text]
  • NICU Gene List Generator.Xlsx
    Neonatal Crisis Sequencing Panel Gene List Genes: A2ML1 - B3GLCT A2ML1 ADAMTS9 ALG1 ARHGEF15 AAAS ADAMTSL2 ALG11 ARHGEF9 AARS1 ADAR ALG12 ARID1A AARS2 ADARB1 ALG13 ARID1B ABAT ADCY6 ALG14 ARID2 ABCA12 ADD3 ALG2 ARL13B ABCA3 ADGRG1 ALG3 ARL6 ABCA4 ADGRV1 ALG6 ARMC9 ABCB11 ADK ALG8 ARPC1B ABCB4 ADNP ALG9 ARSA ABCC6 ADPRS ALK ARSL ABCC8 ADSL ALMS1 ARX ABCC9 AEBP1 ALOX12B ASAH1 ABCD1 AFF3 ALOXE3 ASCC1 ABCD3 AFF4 ALPK3 ASH1L ABCD4 AFG3L2 ALPL ASL ABHD5 AGA ALS2 ASNS ACAD8 AGK ALX3 ASPA ACAD9 AGL ALX4 ASPM ACADM AGPS AMELX ASS1 ACADS AGRN AMER1 ASXL1 ACADSB AGT AMH ASXL3 ACADVL AGTPBP1 AMHR2 ATAD1 ACAN AGTR1 AMN ATL1 ACAT1 AGXT AMPD2 ATM ACE AHCY AMT ATP1A1 ACO2 AHDC1 ANK1 ATP1A2 ACOX1 AHI1 ANK2 ATP1A3 ACP5 AIFM1 ANKH ATP2A1 ACSF3 AIMP1 ANKLE2 ATP5F1A ACTA1 AIMP2 ANKRD11 ATP5F1D ACTA2 AIRE ANKRD26 ATP5F1E ACTB AKAP9 ANTXR2 ATP6V0A2 ACTC1 AKR1D1 AP1S2 ATP6V1B1 ACTG1 AKT2 AP2S1 ATP7A ACTG2 AKT3 AP3B1 ATP8A2 ACTL6B ALAS2 AP3B2 ATP8B1 ACTN1 ALB AP4B1 ATPAF2 ACTN2 ALDH18A1 AP4M1 ATR ACTN4 ALDH1A3 AP4S1 ATRX ACVR1 ALDH3A2 APC AUH ACVRL1 ALDH4A1 APTX AVPR2 ACY1 ALDH5A1 AR B3GALNT2 ADA ALDH6A1 ARFGEF2 B3GALT6 ADAMTS13 ALDH7A1 ARG1 B3GAT3 ADAMTS2 ALDOB ARHGAP31 B3GLCT Updated: 03/15/2021; v.3.6 1 Neonatal Crisis Sequencing Panel Gene List Genes: B4GALT1 - COL11A2 B4GALT1 C1QBP CD3G CHKB B4GALT7 C3 CD40LG CHMP1A B4GAT1 CA2 CD59 CHRNA1 B9D1 CA5A CD70 CHRNB1 B9D2 CACNA1A CD96 CHRND BAAT CACNA1C CDAN1 CHRNE BBIP1 CACNA1D CDC42 CHRNG BBS1 CACNA1E CDH1 CHST14 BBS10 CACNA1F CDH2 CHST3 BBS12 CACNA1G CDK10 CHUK BBS2 CACNA2D2 CDK13 CILK1 BBS4 CACNB2 CDK5RAP2
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • A Dissertation Entitled the Androgen Receptor
    A Dissertation entitled The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit Submitted to the Graduate Faculty as partial fulfillment of the requirements for the PhD Degree in Biomedical science Dr. Manohar Ratnam, Committee Chair Dr. Lirim Shemshedini, Committee Member Dr. Robert Trumbly, Committee Member Dr. Edwin Sanchez, Committee Member Dr. Beata Lecka -Czernik, Committee Member Dr. Patricia R. Komuniecki, Dean College of Graduate Studies The University of Toledo August 2011 Copyright 2011, Mesfin Gonit This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit As partial fulfillment of the requirements for the PhD Degree in Biomedical science The University of Toledo August 2011 Prostate cancer depends on the androgen receptor (AR) for growth and survival even in the absence of androgen. In the classical models of gene activation by AR, ligand activated AR signals through binding to the androgen response elements (AREs) in the target gene promoter/enhancer. In the present study the role of AREs in the androgen- independent transcriptional signaling was investigated using LP50 cells, derived from parental LNCaP cells through extended passage in vitro. LP50 cells reflected the signature gene overexpression profile of advanced clinical prostate tumors. The growth of LP50 cells was profoundly dependent on nuclear localized AR but was independent of androgen. Nevertheless, in these cells AR was unable to bind to AREs in the absence of androgen.
    [Show full text]
  • An All-To-All Approach to the Identification of Sequence-Specific Readers for Epigenetic DNA Modifications on Cytosine
    bioRxiv preprint doi: https://doi.org/10.1101/638700; this version posted May 16, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. An All-to-All Approach to the Identification of Sequence-Specific Readers for Epigenetic DNA Modifications on Cytosine Guang Song1,6, Guohua Wang2,6, Ximei Luo2,3,6, Ying Cheng4, Qifeng Song1, Jun Wan3, Cedric Moore1, Hongjun Song5, Peng Jin4, Jiang Qian3,7,*, Heng Zhu1,7,8,* 1Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 2School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China 3Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 4Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA 5Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA 19104, USA 6These authors contributed equally 7Senior author 8Lead Contact *Correspondence: [email protected] (H.Z.), [email protected] (J.Q.). 1 bioRxiv preprint doi: https://doi.org/10.1101/638700; this version posted May 16, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. SUMMARY Epigenetic modifications of DNA in mammals play important roles in many biological processes. Identification of readers of these epigenetic marks is a critical step towards understanding the underlying molecular mechanisms. Here, we report the invention and application of an all-to-all approach, dubbed Digital Affinity Profiling via Proximity Ligation (DAPPL), to simultaneously profile human TF-DNA interactions using mixtures of random DNA libraries carrying four different epigenetic modifications (i.e., 5-methylcytosine, 5- hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine).
    [Show full text]
  • Developmental Biology 399 (2015) 164–176
    Developmental Biology 399 (2015) 164–176 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology The requirement of histone modification by PRDM12 and Kdm4a for the development of pre-placodal ectoderm and neural crest in Xenopus Shinya Matsukawa a, Kyoko Miwata b, Makoto Asashima b, Tatsuo Michiue a,n a Department of Sciences (Biology), Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan b Research Center for Stem Cell Engineering National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba City, Ibaraki, Japan article info abstract Article history: In vertebrates, pre-placodal ectoderm and neural crest development requires morphogen gradients and Received 6 September 2014 several transcriptional factors, while the involvement of histone modification remains unclear. Here, we Received in revised form report that histone-modifying factors play crucial roles in the development of pre-placodal ectoderm 21 November 2014 and neural crest in Xenopus. During the early neurula stage, PRDM12 was expressed in the lateral pre- Accepted 23 December 2014 placodal ectoderm and repressed the expression of neural crest specifier genes via methylation of Available online 6 January 2015 histone H3K9. ChIP-qPCR analyses indicated that PRDM12 promoted the occupancy of the trimethylated Keywords: histone H3K9 (H3K9me3) on the Foxd3, Slug, and Sox8 promoters. Injection of the PRDM12 MO inhibited fi Histone modi cation the expression of presumptive trigeminal placode markers and decreased the occupancy of H3K9me3 on PRDM12 the Foxd3 promoter. Histone demethylase Kdm4a also inhibited the expression of presumptive Kdm4a trigeminal placode markers in a similar manner to PRDM12 MO and could compensate for the effects Pre-placodal ectoderm Neural crest of PRDM12.
    [Show full text]
  • Downloaded 22.3.2019) Supplemented
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.11.088625; this version posted June 9, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Müller-Eigner et al. 1 Title: Dietary intervention improves health metrics and life expectancy of the genetically obese DU6 (Titan) mouse Authors: Annika Müller-Eigner1†, Adrián Sanz-Moreno2†, Irene de-Diego1†, Anuroop Venkateswaran Venkatasubramani3, Martina Langhammer4, Raffaele Gerlini2,5, Birgit Rathkolb2,5,6, Antonio Aguilar-Pimentel2, Tanja Klein- Rodewald2, Julia Calzada-Wack2, Lore Becker2, Sergio Palma-Vera7, Benedikt Gille1, Ignasi Forne3, Axel Imhof3, Chen Meng8, Christina Ludwig8, Franziska Koch9, Angela Kuhla10, Vanessa Caton11, Julia Brenmoehl11, Jennifer Schoen7,12, Helmut Fuchs2, Valerie Gailus-Durner2, Andreas Hoeflich11, Martin Hrabe de Angelis2,5,13 and Shahaf Peleg1* Affiliations: 1Research Group Epigenetics, Metabolism and Longevity, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany 2German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health (GmbH), 85764 Neuherberg, Germany 3Department of Molecular Biology, Biomedical Center Munich, Ludwig-Maximilians University, 82152 Planegg-Martinsried, Germany 4Institute Genetics and Biometry, Lab Animal Facility, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany 5German Center for Diabetes Research (DZD), 85764,
    [Show full text]