Single-Cell Transcriptional Dynamics and Origins of Neuronal Diversity in the Developing Mouse Neocortex

Total Page:16

File Type:pdf, Size:1020Kb

Single-Cell Transcriptional Dynamics and Origins of Neuronal Diversity in the Developing Mouse Neocortex bioRxiv preprint doi: https://doi.org/10.1101/409458; this version posted September 6, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. Single-cell transcriptional dynamics and origins of neuronal diversity in the developing mouse neocortex L. Telley1,3*†, G. Agirman1,4†, J. Prados1, S. Fièvre1, P. Oberst1, I. Vitali1, L. Nguyen4, A. Dayer1,5, D. Jabaudon1,2* 1 Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland. 2 Clinic of Neurology, Geneva University Hospital, Geneva, Switzerland. 3 Current address: Department of Basic Neuroscience, University of Lausanne, Switzerland. 4 GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège, Belgium. 5 Department of Psychiatry, Geneva University Hospital, Geneva, Switzerland. † equally contributed to this work. * Correspondence to: [email protected]; [email protected] During cortical development, distinct subtypes of glutamatergic neurons are sequentially born and differentiate from dynamic populations of progenitors. The neurogenic competence of these progenitors progresses as corticogenesis proceeds; likewise, newborn neurons transit through sequential states as they differentiate. Here, we trace the developmental transcriptional trajectories of successive generations of apical progenitors (APs) and isochronic cohorts of their daughter neurons using parallel single-cell RNA sequencing between embryonic day (E) 12 and E15 in the mouse cerebral cortex. Our results identify the birthdate- and differentiation stage- related transcriptional dynamics at play during corticogenesis. As corticogenesis proceeds, APs transit through embryonic age-dependent molecular states, which are transmitted to their progeny to generate successive initial daughter cell identities. In neurons, essentially conserved post-mitotic differentiation programs are applied onto these distinct AP-derived ground states, allowing temporally-regulated sequential emergence of specialized neuronal cell types. Molecular temporal patterning of sequentially-born daughter neurons by their respective mother cell thus underlies emergence of neuronal diversity in the neocortex. One Sentence Summary: During corticogenesis, temporally dynamic molecular birthmarks are transmitted from progenitors to their post-mitotic progeny to generate neuronal diversity. The cerebral cortex is a cellularly heterogeneous 2016; Gao et al. 2014, Guo et al. 2013; Gaspard structure, whose neuronal circuits underlie high- et al. 2007); likewise, newborn neurons transit order cognitive and sensorimotor information through sequential transcriptional states as they processing. During embryogenesis, distinct differentiate (Zahr et al. 2018; Telley et al. 2016; subtypes of glutamatergic neurons are Azim et al. 2009). Although the single-cell sequentially born and differentiate from transcriptional diversity of the neocortex is populations of progenitors located in the increasingly well characterized (Saunders et al. germinal zones below the cortex (Jabaudon 2018; Zeisel et al. 2018; Kageyama et al. 2018; 2017; Florio & Huttner 2014). The aggregate Nowakowski et al. 2017; Tasic et al. 2016; neurogenic competence of ventricular zone Zeisel et al. 2015), little is yet known about the progenitors (i.e. apical progenitors, APs) molecular processes driving either the progresses as corticogenesis proceeds progression of AP competence, or the specific (Govindan & Jabaudon 2017; Okamoto et al. -1- bioRxiv preprint doi: https://doi.org/10.1101/409458; this version posted September 6, 2018. 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. differentiation of daughter neurons born from axis, corresponding to the birth and maturation these progenitors at sequential embryonic ages. of daughter neurons, and a birthdate axis, corresponding to the temporal progression in To address these questions, we used AP transcriptional states at sequential FlashTag (FT), a high temporal resolution embryonic ages. These two cardinal processes method to pulse-label APs and their daughter are the major source of transcriptional diversity neurons (Telley et al. 2016; Govindan et al. in the developing neocortex. 2018), to trace the transcriptional trajectories of We used a graph-based cluster analysis successive generations of APs and isochronic to investigate the diversity of differentiation cohorts of their daughter neurons between stage- and birthdate-specific cells and identified embryonic day (E) 12 and E15. This four embryonic age-defined AP transcriptional corresponds to the period during which APs states, as well as two embryonic age-defined successively generate layer (L) 6, L5, L4 and basal progenitor populations, as recently L2/3 neurons (Jabaudon 2017). Following reported (Yuzwa et al. 2017) (Fig. 1C). Two microdissection of the putative somatosensory + classes of 1-day-old neurons (N1d) could be cortex, we collected FT cells by FACS after distinguished, early-born cells (i.e. E12-E13- either 1 h, as APs are still dividing, 24 h, as born) and later-born cells (i.e. E14-E15-born). daughter cells are transiting through the These two classes of neurons displayed early subventricular zone, or 96 h (i.e. four days), onset expression of deep- and superficial-layer once daughter neurons have entered the cortical markers, which foreshadowed their upcoming plate (Fig. 1A and fig. S1, A and B) (Telley et lamina-related identity (Fig. 1C). Classical al. 2016; Govindan et al. 2018). We performed deep-layer markers were also expressed by late- single-cell RNA sequencing at each of these 3 born neurons (fig S2A), consistent with an differentiation stages and 4 embryonic ages initial period of mixed identity followed by (E12, E13, E14, and E15), which yielded a total molecular cross-interactions and progressive of 2,756 quality-controlled cells across 12 fate refinement (Zahr et al. 2018; Ozair et al. conditions for analysis (fig. S1, C and D, and 2018; Azim et al. 2009). Accordingly, by four Methods). days of age, neurons with mutually-exclusive Analysis of cellular transcriptional expression of classical lamina-specific markers identities using t-SNE dimensionality reduction such as Bcl11b (an L5 marker), Rorb (L4) and revealed two main axes of organization: a Cux1 (L2/3) emerged. Of note, GABAergic differentiation (i.e. collection time) axis and a interneurons were also identified (Fig. 1C and birthdate (i.e. injection day) axis (Fig. 1B). fig. S2B), likely corresponding to cells Along the differentiation axis (Fig. 1B, left), 1 migrating into the dorsal pallium after FT h-, 1-day- and 4-day-old cells were organized labeling of their progenitors in the ventral into clusters which corresponded to (1) APs, (2) pallium (Govindan et al. 2018; Wamsley & basal progenitors (BPs) and 1-day-old neurons Fishell 2017; Marin 2013). Astrocytes, (N1d), and (3) 4-day-old neurons (N4d), as corresponding to 4-day-old daughter cells of indicated by the combined expression of type- E15 APs (Minocha et al. 2017; Cahoy et al. specific markers (Telley et al. 2016). Cells born 2008) were also present (Fig. 1C and fig. S2C). at successive times of corticogenesis were These two cell-types were not further organized perpendicularly to this differentiation investigated in this study. Differential axis, forming a birthdate axis (Fig. 1B, right). expression analysis identified type-enriched This chronotopic map was particularly apparent transcripts (Fig. 1D) whose temporal patterns of for APs and 1-day-old daughter cells, but less expression were confirmed using in situ striking in 4-day-old neurons, suggesting that hybridization (Fig. 1E; figs. S3 and S4; ISH; the salience of birthdate-related transcriptional Allen Developing Mouse Brain Atlas). features decreases with differentiation. Together, these results indicate that APs transit Together, these data reveal two orthogonal axes through temporally dynamic transcriptional of transcriptional organization: a differentiation states during corticogenesis as daughter neurons -2- bioRxiv preprint doi: https://doi.org/10.1101/409458; this version posted September 6, 2018. 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. Differentiation Birthdate Collection time Neurod2 Injection day +96h N ABFT 1h E12 +96h 24h Tubb3 E13 96h N4d E14 E15 Eomes N1d BP CP Btg2 +24h BP +24h AP Sox2 FT +1h AP Collection time SVZ Pax6 +1h VZ FT t-SNE2 E12 E13 E14 E15 E12 100 μm t-SNE1 Expression: Min Max Injection day Layer or cell-type C E12 1314 15 marker genes n cells D Enriched genes E E11.5E13.5 E15.5 early 190 mid1 228 early CP E15.5 P4 AP mid2 195 late 230 AP L2/3 1 VZ ly IN r L4 a e N4d early 107 n = 13 ISH L5 BP late 121 CP L6 N4d early 320 mid N1d N1d late 349 n = 13 AP early1 31 E18.5 P4 BP Astro early2 327 n = 6 N4d late1 146 2 L2/3 late2 205 L4 e AP lat L5 Astro 64 late N4d N4d L6 IN.1 142 AP Others IN.2 101 1 150 n = 8 n = 14 200 μm Rorb 0 100 Gfap Cux1 Sox5 Sox2 Gad2 Fezf2 Satb2 Expression (% max): 0 100 Expression: Min Max Bcl11b % total Eomes Neurod2 DL SL Fig. 1. Birthdate and differentiation stage-related cellular diversity in the developing neocortex. (A) Schematic illustration of the experimental procedure. M-phase APs were labeled by FT injection performed at either E12, E13, E14 or E15 and isochronic cohorts of APs and daughter cells were collected either 1, 24 or 96 hours later. (B) t-SNE representation of the single cell RNA sequencing dataset revealing the transcriptional organization of the cells according to the time at which they were collected (i.e. their differentiation status) and the day on which the injection was performed (i.e. their birthdate). APs, BPs and Ns can be distinguished by their combinatorial expression of key marker genes (n = 20 transcripts). (C) Cluster analysis reveals transcriptomically distinct and temporally dynamic cellular clusters. Cluster nomenclature reflects prevalence of the cluster at a given embryonic age (early: E12/E13, late: E14/E15).
Recommended publications
  • A Specific FMNL2 Isoform Is Up-Regulated in Invasive Cells Christine Péladeau, Allan Heibein, Melissa T
    Péladeau et al. BMC Cell Biology (2016) 17:32 DOI 10.1186/s12860-016-0110-z RESEARCH ARTICLE Open Access A specific FMNL2 isoform is up-regulated in invasive cells Christine Péladeau, Allan Heibein, Melissa T. Maltez, Sarah J. Copeland and John W. Copeland* Abstract Background: Formins are a highly conserved family of cytoskeletal remodeling proteins. A growing body of evidence suggests that formins play key roles in the progression and spread of a variety of cancers. There are 15 human formin proteins and of these the Diaphanous-Related Formins (DRFs) are the best characterized. Included in the DRFs are the Formin-Like proteins, FMNL1, 2 & 3, each of which have been strongly implicated in driving tumorigenesis and metastasis of specific tumors. In particular, increased FMNL2 expression correlates with increased invasiveness of colorectal cancer (CRC) in vivo and for a variety of CRC cell-lines in vitro. FMNL2 expression is also required for invasive cell motility in other cancer cell-lines. There are multiple alternatively spliced isoforms of FMNL2 and it is predicted that the encoded proteins will differ in their regulation, subcellular localization and in their ability to regulate cytoskeletal dynamics. Results: Using RT-PCR we identified four FMNL2 isoforms expressed in CRC and melanoma cell-lines. We find that a previously uncharacterized FMNL2 isoform is predominantly expressed in a variety of melanoma and CRC cell lines; this isoform is also more effective in driving 3D motility. Building on previous reports, we also show that FMNL2 is required for invasion in A375 and WM266.4 melanoma cells. Conclusions: Taken together, these results suggest that FMNL2 is likely to be generally required in melanoma cells for invasion, that a specific isoform of FMNL2 is up-regulated in invasive CRC and melanoma cells and this isoform is the most effective at facilitating invasion.
    [Show full text]
  • Snapshot: Formins Christian Baarlink, Dominique Brandt, and Robert Grosse University of Marburg, Marburg 35032, Germany
    SnapShot: Formins Christian Baarlink, Dominique Brandt, and Robert Grosse University of Marburg, Marburg 35032, Germany Formin Regulators Localization Cellular Function Disease Association DIAPH1/DIA1 RhoA, RhoC Cell cortex, Polarized cell migration, microtubule stabilization, Autosomal-dominant nonsyndromic deafness (DFNA1), myeloproliferative (mDia1) phagocytic cup, phagocytosis, axon elongation defects, defects in T lymphocyte traffi cking and proliferation, tumor cell mitotic spindle invasion, defects in natural killer lymphocyte function DIAPH2 Cdc42 Kinetochore Stable microtubule attachment to kinetochore for Premature ovarian failure (mDia3) chromosome alignment DIAPH3 Rif, Cdc42, Filopodia, Filopodia formation, removing the nucleus from Increased chromosomal deletion of gene locus in metastatic tumors (mDia2) Rac, RhoB, endosomes erythroblast, endosome motility, microtubule DIP* stabilization FMNL1 (FRLα) Cdc42 Cell cortex, Phagocytosis, T cell polarity Overexpression is linked to leukemia and non-Hodgkin lymphoma microtubule- organizing center FMNL2/FRL3/ RhoC ND Cell motility Upregulated in metastatic colorectal cancer, chromosomal deletion is FHOD2 associated with mental retardation FMNL3/FRL2 Constituently Stress fi bers ND ND active DAAM1 Dishevelled Cell cortex Planar cell polarity ND DAAM2 ND ND ND Overexpressed in schizophrenia patients Human (Mouse) FHOD1 ROCK Stress fi bers Cell motility FHOD3 ND Nestin, sarcomere Organizing sarcomeres in striated muscle cells Single-nucleotide polymorphisms associated with type 1 diabetes
    [Show full text]
  • Single Nucleotide Polymorphisms in DKK3 Gene Are Associated with Prostate Cancer Risk and Progression ______
    ORIGINAL ARTICLE Vol. 41 (5): 869-897, September - October, 2015 doi: 10.1590/S1677-5538.IBJU.2014.0041 Single nucleotide polymorphisms in DKK3 gene are associated with prostate cancer risk and progression _______________________________________________ Min Su Kim 1, Ha Na Lee 2, Hae Jong Kim 3,4, Soon Chul Myung 5 1Department of Urology, Seoul Medical Center, Seoul, Korea; 2Department of Urology, Seoul Seonam Hospital, EwhaWomans University, Seoul, Korea; 3Research Institue for Biomedical and Pharmaceutical Sciences, Chung-Ang University, Seoul, Korea; 4Advanced Urogenital Diseas Research Center, Chung-Ang University, College of Medicine, Seoul, Korea; 5Department of Urology, Chung-Ang University, College of Medicine, Seoul, Korea ABSTRACT ARTICLE INFO ______________________________________________________________ ______________________ We had investigated whether sequence variants within DKK3 gene are associated with Key words: the development of prostate cancer in a Korean study cohort. We evaluated the asso- Biological Markers; Genetic ciation between 53 single nucleotide polymorphisms (SNPs) in the DKK3 gene and Variation; Prostatic Neoplasms; prostate cancer risk as well as clinical characteristics (PSA, clinical stage, pathological Polymorphism, Single Nucleotide stage and Gleason score) in Korean men (272 prostate cancer subjects and 173 benign prostate hyperplasia subjects) using unconditional logistic regression analysis. Of the Int Braz J Urol. 2015; 41: 869-97 53 SNPs and 25 common haplotypes, 5 SNPs and 4 haplotypes were
    [Show full text]
  • Short Bowel Syndrome Results in Increased Gene Expression
    Schall et al. BMC Genomics (2017) 18:23 DOI 10.1186/s12864-016-3433-4 RESEARCH ARTICLE Open Access Short bowel syndrome results in increased gene expression associated with proliferation, inflammation, bile acid synthesis and immune system activation: RNA sequencing a zebrafish SBS model Kathy A. Schall1, Matthew E. Thornton2, Mubina Isani1, Kathleen A. Holoyda1, Xiaogang Hou1, Ching-Ling Lien3, Brendan H. Grubbs2 and Tracy C. Grikscheit1,4* Abstract Background: Much of the morbidity associated with short bowel syndrome (SBS) is attributed to effects of decreased enteral nutrition and administration of total parenteral nutrition (TPN). We hypothesized that acute SBS alone has significant effects on gene expression beyond epithelial proliferation, and tested this in a zebrafish SBS model. Methods: In a model of SBS in zebrafish (laparotomy, proximal stoma, distal ligation, n = 29) or sham (laparotomy alone, n = 28) surgery, RNA-Seq was performed after 2 weeks. The proximal intestine was harvested and RNA isolated. The three samples from each group with the highest amount of RNA were spiked with external RNA controls consortium (ERCC) controls, sequenced and aligned to reference genome with gene ontology (GO) enrichment analysis performed. Gene expression of ctnnb1, ccnb1, ccnd1, cyp7a1a, dkk3, ifng1-2, igf2a, il1b, lef1, nos2b, saa1, stat3, tnfa and wnt5a were confirmed to be elevated in SBS by RT-qPCR. Results: RNA-seq analysis identified 1346 significantly upregulated genes and 678 significantly downregulated genes in SBS zebrafish intestine compared to sham with Ingenuity analysis. The upregulated genes were involved in cell proliferation, acute phase response signaling, innate and adaptive immunity, bile acid regulation, production of nitric oxide and reactive oxygen species, cellular barrier and coagulation.
    [Show full text]
  • Not Dicer but Argonaute Is Required for a Microrna Production
    Cell Research (2010) 20:735-737. npg © 2010 IBCB, SIBS, CAS All rights reserved 1001-0602/10 $ 32.00 RESEARCH HIGHLIGHT www.nature.com/cr A new twist in the microRNA pathway: Not Dicer but Argonaute is required for a microRNA production Gabriel D Bossé1, Martin J Simard1 1Laval University Cancer Research Centre, Hôtel-Dieu de Québec (CHUQ), Quebec City, Québec G1R 2J6, Canada Cell Research (2010) 20:735-737. doi:10.1038/cr.2010.83; published online 15 June 2010 Found in all metazoans, microRNAs A Canonical pathway B Ago2-dependent pathway or miRNAs are small non-coding RNA Nucleus Cytoplasm Nucleus Cytoplasm of ~22 nucleotides in length that com- Exp.5 Exp.5 pletely reshaped our understanding of gene regulation. This new class of gene pre-miR-451 regulator is mostly transcribed by the pre-miRNA RNA polymerase II producing a long stem-loop structure, called primary- or Ago2 pri-miRNA, that will first be processed Ago2 Dicer in the cell nucleus by a multiprotein TRBP complex called microprocessor to gen- erate a shorter RNA structure called Ago2 RISC precursor- or pre-miRNA. The precisely Ago2 RISC processed pre-miRNA will next be ex- ported into the cytoplasm by Exportin 5 and loaded onto another processing machine containing the ribonuclease III enzyme Dicer, an Argonaute protein Ago2 Ago2 and other accessory cellular factors mRNA mRNA (Figure 1A; [1]). Dicer will mediate the Translation inhibition Translation inhibition cleavage of the pre-miRNA to form the mature miRNA that will then be bound Figure 1 (A) Canonical microRNA biogenesis. In mammals, the pre-miRNA is by the Argonaute protein to form, most loaded onto a multiprotein complex consisting minimally of Dicer, Tar RNA Bind- likely with other cellular factors, the ef- ing Protein (TRBP) and Ago2.
    [Show full text]
  • EIF2C2 Monoclonal Antibody (M01), Clone 2E12-1C9
    EIF2C2 monoclonal antibody (M01), clone 2E12-1C9 Catalog # : H00027161-M01 規格 : [ 100 ug ] List All Specification Application Image Product Mouse monoclonal antibody raised against a full length recombinant Western Blot (Cell lysate) Description: EIF2C2. Immunogen: EIF2C2 (AAH07633.1, 483 a.a. ~ 859 a.a) full-length recombinant protein with GST tag. MW of the GST tag alone is 26 KDa. Sequence: MPIQGQPCFCKYAQGADSVEPMFRHLKNTYAGLQLVVVILPGKTPVYAE VKRVGDTVLGMATQCVQMKNVQRTTPQTLSNLCLKINVKLGGVNNILLP enlarge QGRPPVFQQPVIFLGADVTHPPAGDGKKPSIAAVVGSMDAHPNRYCATV Western Blot (Transfected RVQQHRQEIIQDLAAMVRELLIQFYKSTRFKPTRIIFYRDGVSEGQFQQV lysate) LHHELLAIREACIKLEKDYQPGITFIVVQKRHHTRLFCTDKNERVGKSGNIP AGTTVDTKITHPTEFDFYLCSHAGIQGTSRPSHYHVLWDDNRFSSDELQI LTYQLCHTYVRCTRSVSIPAPAYYAHLVAFRARYHLVDKEHDSAEGSHTS GQSNGRDHQALAKAVQVHQDTLRTMYFA Host: Mouse enlarge Reactivity: Human Western Blot (Recombinant Isotype: IgG1 Kappa protein) Quality Control Antibody Reactive Against Recombinant Protein. Immunofluorescence Testing: enlarge Immunohistochemistry (Formalin/PFA-fixed paraffin- embedded sections) Western Blot detection against Immunogen (68.03 KDa) . Storage Buffer: In 1x PBS, pH 7.4 enlarge Storage Store at -20°C or lower. Aliquot to avoid repeated freezing and thawing. Instruction: Sandwich ELISA (Recombinant protein) MSDS: Download Interspecies Mouse (100); Rat (99) Antigen Sequence: enlarge Datasheet: Download ELISA Publication Reference RNAi Knockdown (Antibody validated) 1. TDP-43 aggregation induced by oxidative stress causes global mitochondrial imbalance
    [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]
  • Noelia Díaz Blanco
    Effects of environmental factors on the gonadal transcriptome of European sea bass (Dicentrarchus labrax), juvenile growth and sex ratios Noelia Díaz Blanco Ph.D. thesis 2014 Submitted in partial fulfillment of the requirements for the Ph.D. degree from the Universitat Pompeu Fabra (UPF). This work has been carried out at the Group of Biology of Reproduction (GBR), at the Department of Renewable Marine Resources of the Institute of Marine Sciences (ICM-CSIC). Thesis supervisor: Dr. Francesc Piferrer Professor d’Investigació Institut de Ciències del Mar (ICM-CSIC) i ii A mis padres A Xavi iii iv Acknowledgements This thesis has been made possible by the support of many people who in one way or another, many times unknowingly, gave me the strength to overcome this "long and winding road". First of all, I would like to thank my supervisor, Dr. Francesc Piferrer, for his patience, guidance and wise advice throughout all this Ph.D. experience. But above all, for the trust he placed on me almost seven years ago when he offered me the opportunity to be part of his team. Thanks also for teaching me how to question always everything, for sharing with me your enthusiasm for science and for giving me the opportunity of learning from you by participating in many projects, collaborations and scientific meetings. I am also thankful to my colleagues (former and present Group of Biology of Reproduction members) for your support and encouragement throughout this journey. To the “exGBRs”, thanks for helping me with my first steps into this world. Working as an undergrad with you Dr.
    [Show full text]
  • WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization I International Bureau (10) International Publication Number (43) International Publication Date WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT (51) International Patent Classification: CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, C12Q 1/68 (2018.01) A61P 31/18 (2006.01) DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, C12Q 1/70 (2006.01) HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, (21) International Application Number: MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, PCT/US2018/056167 OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, (22) International Filing Date: SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, 16 October 2018 (16. 10.2018) TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every kind of regional protection available): ARIPO (BW, GH, (26) Publication Language: English GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, (30) Priority Data: UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, 62/573,025 16 October 2017 (16. 10.2017) US TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, FR, GB, GR, HR, HU, ΓΕ , IS, IT, LT, LU, LV, (71) Applicant: MASSACHUSETTS INSTITUTE OF MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, TECHNOLOGY [US/US]; 77 Massachusetts Avenue, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, Cambridge, Massachusetts 02139 (US).
    [Show full text]
  • A Rhoa and Rnd3 Cycle Regulates Actin Reassembly During Membrane
    A RhoA and Rnd3 cycle regulates actin reassembly PNAS PLUS during membrane blebbing Kana Aokia, Fumiyo Maedaa,1, Tomoya Nagasakob,1, Yuki Mochizukia, Seiichi Uchidab, and Junichi Ikenouchia,c,d,2 aDepartment of Biology, Faculty of Sciences, Kyushu University, Fukuoka 819-0395, Japan; bDepartment of Advanced Information Technology, Kyushu University, Fukuoka 819-0395, Japan; cPrecursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan; and dAMED-PRIME, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan Edited by Thomas D. Pollard, Yale University, New Haven, CT, and approved February 12, 2016 (received for review January 21, 2016) The actin cytoskeleton usually lies beneath the plasma membrane. membrane blebs is promoted by epidermal growth factor receptor When the membrane-associated actin cytoskeleton is transiently kinase substrate 8 (Eps8) and ezrin, and regulated by a RhoA– disrupted or the intracellular pressure is increased, the plasma Rho-associated protein kinase (ROCK)–Rnd3 feedback loop. membrane detaches from the cortex and protrudes. Such protruded membrane regions are called blebs. However, the molecular mech- Results and Discussion anisms underlying membrane blebbing are poorly understood. This Membrane Blebs Retract from Multiple Sites. We used the human study revealed that epidermal growth factor receptor kinase sub- colon carcinoma cell line DLD1 to observe membrane blebbing. strate 8 (Eps8) and ezrin are important regulators of rapid actin When cultured in 2D conditions, DLD1 cells did not exhibit reassembly for the initiation and retraction of protruded blebs. Live- membrane blebbing (Fig. 1A, Left). However, DLD1 cells ac- cell imaging of membrane blebbing revealed that local reassembly tively formed membrane blebs when embedded in a type I col- of actin filaments occurred at Eps8- and activated ezrin-positive foci lagen gel (Fig.
    [Show full text]