Supporting Information

Total Page:16

File Type:pdf, Size:1020Kb

Supporting Information Supporting Information McKenna et al. 10.1073/pnas.1504144112 SI Results Primary antibodies used in this study were as follows: chicken Corticothalamic Axons Are Present in the Satb2 cKO Mice. The ex- anti–β-gal (ab9361; Abcam), 1:1,000; rat anti-CTIP2 (ab18465; pression of SATB2 in corticothalamic neurons (Fig. 1 E–H and J) Abcam), 1:200; rabbit anti-CUX1 (sc-13024; Santa Cruz Bio- suggests that it may regulate the development of corticothalamic technology), 1:500; rabbit anti-DARPP32 (ab40801; Abcam), neurons and axons. To examine the corticofugal axons in Satb2 1:200; goat anti-BHLHB5 (sc-6045; Santa Cruz Biotechnology), cKO mice, we used two genetic markers to specifically label PLAP 1:100; rabbit anti-FOG2 (ZFMP2; sc-10755; Santa Cruz Bio- these axons. The Fezf2 allele contains a human PLAP technology), FOXP2 (ab16046; Abcam), 1:1,000; rabbit anti- Fezf2 cDNA that was inserted in the gene, and labels cortico- GFP (A11122; Invitrogen), 1:1,000; rabbit anti-NFIA (39397; Golli-GFP thalamic and subcerebral axons with PLAP (6). The Active Motif), 1:500; rabbit anti-PLAP (AHP537; Accurate transgene specifically labels corticothalamic and some sub- Chemical), 1:100; rabbit anti-SATB2 (ab34735; Abcam), 1:1,000; cerebral axons (37). lacZ/+ PLAP/+ mouse anti-SATB2 (sc-81376; Santa Cruz Biotechnology), 1:1,000; Compared with control Satb2 ; Fezf2 mice, we found – and rabbit anti-TBR1 (ab31940; Abcam), 1:500. that more Satb2-LacZ labeled axons were present in the thala- + Emx1-Cre Satb2+/+ enhancer Emx1-Cre Satb2lacZ/flox Fezf2PLAP/+ Satb2 cKO For counting of LacZ cells in the ; ; mus of the P4 ; ; ( ) 434-lacZ Emx1-Cre Satb2flox/flox enhancer-434 Satb2 brains (Fig. S6 A and B). This result is consistent with a recent (control) and ; ; ( cKO n = study (25). Intriguingly, fewer Fezf2-PLAP labeled corticothalamic )cortices( 4 for each genotype, collected from three litters), lacZ/flox PLAP/+ axons were present in the P4 Emx1-Cre; Satb2 ; Fezf2 three sections stained with X-Gal were counted from each animal. + + brains (Fig. S6 C and D). Similarly, reduction of Golli-GFP cor- The quantification for the control genotype was 59 ± 8.2 LacZ lacZ/flox ticothalamic axons was observed in P4 Emx1-Cre; Satb2 ; neurons per square millimeter (mean ± SD), and for the Satb2 cKO + Golli-GFP brains. was 23 ± 9.0 LacZ neurons per square millimeter. To determine To further investigate the corticothalamic axons, we compared statistical significance, GraphPad was used to preform an un- + +/+ the GFP axons in Emx1-Cre; Satb2 ; RCE-GFP (control) and paired t test between experimental groups. lacZ/flox Emx1-Cre; Satb2 ; RCE-GFP (Satb2 cKO) brains. Although + GFP corticospinal axons were absent in the Satb2 cKO mice, a Microscopy. Bright-field and epifluorescence images were taken + +/+ careful comparison of GFP axons between Emx1-Cre; Satb2 ; on an Olympus BX51 microscope by using a Q Imaging Retiga lacZ/flox RCE-GFP and Emx1-Cre; Satb2 ; RCE-GFP brains at P24 EXj camera or a Keyence BZ-9000 microscope. Confocal images did not show a significant difference in the corticothalamic in- were obtained on a Leica TCS SP5 or a Zeiss LSM5 confocal nervation patterns. microscope. Images were processed by using Adobe Photoshop E–P Puzzled by this, we investigated Golli-GFP (Fig. S6 ) and CS5 to adjust brightness and contrast. Fezf2-PLAP (Fig. 4) expression in the cortex, and discovered that these genetic markers were no longer expressed in layer 5 Luciferase Assay. Enhancer 434 was amplified from mouse geno- and upper layer 6 neurons in the Satb2 mutant mice. In the mean mic DNA using primers 5′-ATCGCTCGAGCAGGCTGTAGG- time, we observed complementary expanded expression domain ATGGGCAGCAGGAGTTTC-3′ and 5′-ATCGAAGCTTGTA- of the Satb2-LacZ reporter in the deep layer neurons (Fig. 4 and ′ + + ACAAGTCAGGTGAGCAGGCGGTA-3 . The amplified DNA Fig. S6). A comparison of Golli-GFP and Satb2-LacZ axons Satb2lacZ/+ Golli-GFP Emx1-Cre Satb2lacZ/flox was inserted into the SalI site of the luciferase reporter plasmid between P28 ; and ; ; pGL4CP-TK (29) in both orientations. Thus, the enhancer 434 Golli-GFP mice showed that, although some thalamic nuclei + was inserted downstream of the polyadenylation site of the lu- received less innervation from Golli-GFP axons, more Satb2- + LacZ axons were observed in these nuclei, so that the total ciferase gene. + + The SATB2 binding site in the intron of Sox5 was amplified by innervation pattern by the Golli-GFP and Satb2-LacZ axons ′ ′ was not significantly affected in the Satb2 cKO mice (Fig. S6 E– using primers 5 -ATCCACAATGGTCCGCGGGTAACCG-3 + ′ ′ P). The reduced innervation by Golli-GFP axons was most and 5 -TGTAGGAGGTGCCACAGTGCTGGG-3 . The ampli- apparent in the thalamic nuclei targeted by layer 5 cortico- fied sequence was inserted into the BsrG1 sites of the pGL4CP-TK + thalamic axons (Fig. S6 E–P) (46). More Satb2-LacZ cortico- plasmid in both orientations, upstream of the HSV thymidine ki- thalamic axons were observed in these same nuclei (Fig. S6 E–P). nase gene promoter, which drives luciferase gene expression. Thus, the corticothalamic axonal defects observed in the Satb2 Neuro-2A cells (American Type Culture Collection) were mutant brains using Golli-GFP, Fezf2-PLAP and Satb2-LacZ transfected with pCAG-Satb2 (gift from Nenad Sestan, Yale axonal reporters were at least partially attributable to the changed University, New Haven, CT) or empty pCAG vector, Renilla reporter expressions. Overall, our axonal analysis suggested that luciferase (pRL-SV40) plasmids (internal control), and empty Satb2 has a more limited role in regulating the development of pGL4CP-TK firefly luciferase reporter plasmids, or pGL4CP-TK corticothalamic axons. reporter plasmids containing enhancer 434 or SATB2 binding site in the Sox5 gene, using Lipofectamine 2000 (Invitrogen). SI Methods β Luciferase activities were assayed 48 h later using the Dual- Retrograde Tracing. The fluorescent tracer CT conjugated with Luciferase System (Promega). Three technical replicates were mea- Alexa 488 or Alexa 594 was injected into the thalamus or pyra- sured for each biological replicate. The ratio of firefly luciferase and midal decussation by using Vevo770 ultrasound-guided injection Renillaluciferaseforeachreplicatewascalculated.InFigs.2E and systems on E17.5 or P2. The brains were collected on P2 or P8 I E and processed for immunohistochemistry. 3 , relative luciferase activities for enhancer 434 (Fig. 2 ) and SATB2 binding site in Sox5 (Fig. 3I) in the presence of Satb2 cDNA are Histochemistry. Immunohistochemistry and PLAP staining for mouse shown relative to the activity in the empty pGL4CP-TK plasmid tissues were performed as previously described (9, 39). LacZ staining measured in the presence of Satb2 cDNA. Error bars represent SD. was performed as previously described (15). All experiments were repeated three times. McKenna et al. www.pnas.org/cgi/content/short/1504144112 1of8 Fig. S1. (A–C) LacZ staining shows that Satb2-LacZ+ axons are present in the corpus callosum (red star, A), internal capsule (red arrow, A), cerebral peduncle (red arrow, C), and thalamus (red arrow, B). LacZ staining in the cerebral peduncle and thalamus is less intense than in the corpus callosum. (D–K)Im- munostaining shows that the rabbit SATB2 antibody (Abcam) is specific to the SATB2 protein. (D) SATB2 immunostaining of a P0 WT cortex. (E) No SATB2 lacZ/lacZ flox/+ antibody staining was detected in the Satb2 mice at P0. (F) Low-magnification DAPI image of the control (Satb2 ) brain section shown in G and H. (G) SATB2 immunostaining of the section in F.(H) Higher-magnification image of the boxed area in F and G shows SATB2 staining. (I) DAPI staining of a Satb2 flox/flox cKO (Satb2 ; Emx1-Cre) brain section. (J) SATB2 staining of the same brain section shown in I.(K) Higher-magnification image of the boxed area in I and J shows SATB2 staining. (Scale bars: A–C, 500 μm; D and E, 100 μm; F and I,500μm; G and J, 500 μm; H and K, 100 μm.) McKenna et al. www.pnas.org/cgi/content/short/1504144112 2of8 Fig. S2. Expressions of upper-layer neuronal genes and deep-layer neuronal genes were misregulated in Satb2 mutant mice. (A) RNA-seq analysis showed that lacZ/lacZ +/+ 3,129 genes were misregulated (adjusted P < 0.1) in the P0 Satb2 cortices. Compared with Satb2 cortices, at least 15 upper-layer genes (red tick marks) lacZ/lacZ showed reduced expression, and at least 74 deep-layer genes (blue tick marks) showed increased or decreased expression in the Satb2 cortices. lacZ/lacZ +/+ lacZ/lacZ (B–E) TLE4 expression is reduced in the P0 Satb2 cortices. (B and C) Satb2 brains. (D and E) Satb2 brains. (F–I) NFIA expression is reduced in the P0 Satb2lacZ/lacZ brains. (F and G) Satb2+/+ brains. (H and I) Satb2lacZ/lacZ brains. (J and K) DARPP32 expression was reduced in the Satb2lacZ/lacZ brains. (J) DARPP32 staining in Satb2lacZ/+ cortices. (K) Satb2lacZ/lacZ cortices. (Scale bars: B–E,100μm; F–I, 100 μm; J and K,500μm.) McKenna et al. www.pnas.org/cgi/content/short/1504144112 3of8 Fig. S3. SATB2 regulates Fezf2 expression. (A–J) In situ hybridization showed that Fezf2 mRNA expression was reduced in the E16.5 (A–F) and P0 (G–J) lacZ/lacZ +/+ lacZ/lacZ Satb2 cortices. (A–C, G, and H) Satb2 brains. (D–F, I, and J) Satb2 brains. (K) Immunohistochemistry revealed that β-gal was expressed in cortical +/+ + flox/flox neurons in the P0 control Emx1-Cre; Satb2 ; enhancer 434-lacZ mice. (L) The number of β-gal neurons was reduced in P0 Emx1-Cre; Satb2 ; enhancer +/+ + 434-lacZ mice. (M) Immunohistochemistry for β-gal in the P4 control Emx1-Cre; Satb2 ; enhancer 434-lacZ mice. (N) The number of β-gal neurons was re- flox/flox duced in P4 Emx1-Cre; Satb2 ; enhancer 434-lacZ mice.
Recommended publications
  • 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]
  • The Role of Gli3 in Inflammation
    University of New Hampshire University of New Hampshire Scholars' Repository Doctoral Dissertations Student Scholarship Winter 2020 THE ROLE OF GLI3 IN INFLAMMATION Stephan Josef Matissek University of New Hampshire, Durham Follow this and additional works at: https://scholars.unh.edu/dissertation Recommended Citation Matissek, Stephan Josef, "THE ROLE OF GLI3 IN INFLAMMATION" (2020). Doctoral Dissertations. 2552. https://scholars.unh.edu/dissertation/2552 This Dissertation is brought to you for free and open access by the Student Scholarship at University of New Hampshire Scholars' Repository. It has been accepted for inclusion in Doctoral Dissertations by an authorized administrator of University of New Hampshire Scholars' Repository. For more information, please contact [email protected]. THE ROLE OF GLI3 IN INFLAMMATION BY STEPHAN JOSEF MATISSEK B.S. in Pharmaceutical Biotechnology, Biberach University of Applied Sciences, Germany, 2014 DISSERTATION Submitted to the University of New Hampshire in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy In Biochemistry December 2020 This dissertation was examined and approved in partial fulfillment of the requirement for the degree of Doctor of Philosophy in Biochemistry by: Dissertation Director, Sherine F. Elsawa, Associate Professor Linda S. Yasui, Associate Professor, Northern Illinois University Paul Tsang, Professor Xuanmao Chen, Assistant Professor Don Wojchowski, Professor On October 14th, 2020 ii ACKNOWLEDGEMENTS First, I want to express my absolute gratitude to my advisor Dr. Sherine Elsawa. Without her help, incredible scientific knowledge and amazing guidance I would not have been able to achieve what I did. It was her encouragement and believe in me that made me overcome any scientific struggles and strengthened my self-esteem as a human being and as a scientist.
    [Show full text]
  • Emx2 Is Required for Growth of the Hippocampus but Not for Hippocampal Field Specification
    The Journal of Neuroscience, April 1, 2000, 20(7):2618–2625 Emx2 Is Required for Growth of the Hippocampus But Not for Hippocampal Field Specification Shubha Tole,1 Guy Goudreau,2 Stavroula Assimacopoulos,1 and Elizabeth A. Grove1 1Department of Neurobiology, Pharmacology, and Physiology, University of Chicago, Chicago, Illinois 60637, and 2Max Planck Institute of Biophysical Chemistry, D-37077 Goettingen, Germany The vertebrate Emx genes are expressed in a nested pattern in positioned in the Emx2 mutant. In particular, a dentate cell early embryonic cerebral cortex, such that a medial strip of population is generated, although it fails to form a morpholog- cortex expresses Emx2 but not Emx1. This pattern suggests ical gyrus. This failure may be part of a more widespread medial that Emx genes could play a role in specifying different areas or cortical defect in the mutant. Examination of cortical cell pro- fields of the cortex along the mediolateral axis. Such a role has liferation and differentiation indicates a disruption of the matu- been supported by the observation that in mice lacking func- ration of the medial cortex in the absence of Emx2. Thus, Emx2 tional Emx2 the hippocampus is shrunken and the most medial is required for normal growth and maturation of the hippocam- field of the cortex, the hippocampal dentate gyrus, appears by pus but not for the specification of cells to particular hippocam- cytoarchitecture to be missing (Pellegrini et al., 1996; Yoshida et pal field identities. al., 1997). Use of region-specific molecular markers shows, Key words: Emx2; hippocampus; patterning; specification; however, that hippocampal fields are specified and correctly cortical maturation; cortical hem The hippocampus, like the rest of the cerebral cortex, is divided Reports that specific mutations lead to morphological defects in into cytoarchitectonic areas or fields (Nauta and Feirtag, 1986).
    [Show full text]
  • CIC Is a Critical Regulator of Neuronal Differentiation
    CIC is a critical regulator of neuronal differentiation Inah Hwang, … , Hongwu Zheng, Jihye Paik JCI Insight. 2020. https://doi.org/10.1172/jci.insight.135826. Research In-Press Preview Oncology Stem cells Capicua (CIC), a member of the high mobility group (HMG)-box superfamily of transcriptional repressors, is frequently mutated in human oligodendrogliomas. But its function in brain development and tumorigenesis remains poorly understood. Here, we report that brain-specific deletion of Cic compromises developmental transition of neuroblast to immature neurons in mouse hippocampus and compromises normal neuronal differentiation. Combined gene expression and ChIP-seq analyses identified VGF as an important CIC-repressed transcriptional surrogate involved in neuronal lineage regulation. Aberrant VGF expression promotes neural progenitor cell proliferation by suppressing their differentiation. Mechanistically, we demonstrated that CIC represses VGF expression by tethering SIN3-HDAC to form a transcriptional corepressor complex. Mass spectrometry analysis of CIC-interacting proteins further identified BRG1 containing mSWI/SNF complex whose function is necessary for transcriptional repression by CIC. Together, this study uncovers a novel regulatory pathway of CIC-dependent neuronal differentiation and may implicate these molecular mechanisms in CIC-dependent brain tumorigenesis. Find the latest version: https://jci.me/135826/pdf CIC is a Critical Regulator of Neuronal Differentiation Inah Hwang1,4, Heng Pan2,3,4, Jun Yao5, Olivier Elemento2,3,4, Hongwu Zheng1,4 and Jihye Paik1,4* 1Department of Pathology and Laboratory medicine. Weil Cornell Medicine, New York, NY10021, USA 2Department of Physiology and Biophysics, Weil Cornell Medicine, New York, NY10021, USA 3Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.
    [Show full text]
  • The Role of Emx1 and Emx2 in the Developing Chick Telencephalon
    The Role of Emx1 and Emx2 in the developing chick telencephalon Dissertation Zur Erlangung des Doktorgrades der Naturwissenschaften (Dr.rer.nat.) der Fakultät für Biologie der Ludwig-Maximilian-Universität München Angefertigt am Max-Planck-Institut für Neurobiologie in der Arbeitsgruppe Neuronale Spezifizierung und in der GSF am Institut für Stammzellforschung Julia von Frowein München, Dezember 2004 1. Gutachter: Prof.Dr. Magdalena Götz 2. Gutachter: Prof.Dr. George Boyan eingereicht am 20.12.2004 Tag der mündlichen Prüfung: 26.4.2005 If the brain were so simple we could understand it, we would be so simple we couldn't. Lyall Watson Table of content 1 Table of content 1 Table of content...........................................................................................1 2 Abstract ........................................................................................................5 3 Zusammenfassung......................................................................................6 4 Introduction..................................................................................................8 4.1 General development of the regions of the central nervous system ....................................8 4.2 Patterning and regionalization .............................................................................................8 4.3 Regions of the forebrain.....................................................................................................10 4.4 Migration............................................................................................................................12
    [Show full text]
  • Cortical Foxp2 Supports Behavioral Flexibility and Developmental Dopamine D1 Receptor Expression
    bioRxiv preprint doi: https://doi.org/10.1101/624973; this version posted May 2, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Cortical Foxp2 supports behavioral flexibility and developmental dopamine D1 receptor expression Marissa Co, Stephanie L. Hickey, Ashwinikumar Kulkarni, Matthew Harper, Genevieve Konopka* Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA *Correspondence: [email protected] Contact information Genevieve Konopka, Ph.D. Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., ND4.300, Dallas, TX 75390-9111 TEL: 214-648-5135, FAX: 214-648-1801, Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/624973; this version posted May 2, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract FoxP2 encodes a forkhead box transcription factor required for the development of neural circuits underlying language, vocalization, and motor-skill learning. Human genetic studies have associated FOXP2 variation with neurodevelopmental disorders (NDDs), and within the cortex, it is coexpressed and interacts with other NDD-associated transcription factors. Cortical Foxp2 is required in mice for proper social interactions, but its role in other NDD-relevant behaviors and molecular pathways is unknown.
    [Show full text]
  • New Functions of Long Noncoding Rnas During EMT and Tumor Progression Chunlai Li
    Cancer Cancer Research Highlights Research New Functions of Long Noncoding RNAs during EMT and Tumor Progression Chunlai Li Epithelial-to-mesenchymal transition (EMT) is orches- dysregulated in colorectal carcinoma and correlates with trated by aberrant activation of EMT transcription factors poor survival and prognosis. They present a new functional and expression of downstream target genes resulting in mechanism illustrating how the SATB2-AS1–SATB2–Snail increased tumor initiation, metastatic potential, and thera- axis is involved in epigenetic modification that regulates peutic resistance. In this issue of Cancer Research,Wangand colorectal carcinoma progression. colleagues show that the long noncoding RNA SATB2-AS1 is See related article by Wang et al., p. 3542 The epithelial-to-mesenchymal transition (EMT) is a dynamic plasm, nucleus, or nucleolus. The distinct cellular localization of a process, in which epithelial cells acquire mesenchymal features. It lncRNA determines its molecular function: cytoplasmic lncRNAs is characterized by loss of intercellular contacts and cellular are responsible for posttranscriptional regulation including polarity, and acquisition of motility and invasive properties (1). mRNA stability and protein modification (6); nuclear lncRNAs EMT is crucial during embryonic development, wound healing, have functions at the transcriptional level such as histone mod- tissue regeneration, and organ fibrosis. In addition to the funda- ification, alternative splicing, or direct transcriptional regula- mental role EMT
    [Show full text]
  • SATB2 Induction of a Neural Crest Mesenchyme-Like Program Drives Melanoma Invasion and Drug Resistance
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2-2-2021 SATB2 induction of a neural crest mesenchyme-like program drives melanoma invasion and drug resistance Maurizio Fazio Charles K. Kaufman et al Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs RESEARCH ARTICLE SATB2 induction of a neural crest mesenchyme-like program drives melanoma invasion and drug resistance Maurizio Fazio1,2†, Ellen van Rooijen1,2†, Michelle Dang1,2, Glenn van de Hoek1, Julien Ablain1,2, Jeffrey K Mito1,3, Song Yang1, Andrew Thomas1, Jonathan Michael1, Tania Fabo1,2, Rodsy Modhurima1,2, Patrizia Pessina4, Charles K Kaufman5,6, Yi Zhou1,2, Richard M White7, Leonard I Zon1,2* 1Howard Hughes Medical Institute, Stem Cell Program and the Division of Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States; 2Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, United States; 3Brigham and Women’s Hospital, Department of Pathology, Boston, United States; 4Stem Cell Program and the Division of Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States; 5Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, United States; 6Department of Developmental Biology, Washington University in Saint Louis, St. Louis, United States; 7Memorial Sloan Kettering Cancer Center and Weill-Cornell Medical College, New York, United States Abstract Recent genomic and scRNA-seq analyses of melanoma demonstrated a lack of recurrent genetic drivers of metastasis, while identifying common transcriptional states correlating *For correspondence: with invasion or drug resistance.
    [Show full text]
  • Figure S1. Reverse Transcription‑Quantitative PCR Analysis of ETV5 Mrna Expression Levels in Parental and ETV5 Stable Transfectants
    Figure S1. Reverse transcription‑quantitative PCR analysis of ETV5 mRNA expression levels in parental and ETV5 stable transfectants. (A) Hec1a and Hec1a‑ETV5 EC cell lines; (B) Ishikawa and Ishikawa‑ETV5 EC cell lines. **P<0.005, unpaired Student's t‑test. EC, endometrial cancer; ETV5, ETS variant transcription factor 5. Figure S2. Survival analysis of sample clusters 1‑4. Kaplan Meier graphs for (A) recurrence‑free and (B) overall survival. Survival curves were constructed using the Kaplan‑Meier method, and differences between sample cluster curves were analyzed by log‑rank test. Figure S3. ROC analysis of hub genes. For each gene, ROC curve (left) and mRNA expression levels (right) in control (n=35) and tumor (n=545) samples from The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer cohort are shown. mRNA levels are expressed as Log2(x+1), where ‘x’ is the RSEM normalized expression value. ROC, receiver operating characteristic. Table SI. Clinicopathological characteristics of the GSE17025 dataset. Characteristic n % Atrophic endometrium 12 (postmenopausal) (Control group) Tumor stage I 91 100 Histology Endometrioid adenocarcinoma 79 86.81 Papillary serous 12 13.19 Histological grade Grade 1 30 32.97 Grade 2 36 39.56 Grade 3 25 27.47 Myometrial invasiona Superficial (<50%) 67 74.44 Deep (>50%) 23 25.56 aMyometrial invasion information was available for 90 of 91 tumor samples. Table SII. Clinicopathological characteristics of The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer dataset. Characteristic n % Solid tissue normal 16 Tumor samples Stagea I 226 68.278 II 19 5.740 III 70 21.148 IV 16 4.834 Histology Endometrioid 271 81.381 Mixed 10 3.003 Serous 52 15.616 Histological grade Grade 1 78 23.423 Grade 2 91 27.327 Grade 3 164 49.249 Molecular subtypeb POLE 17 7.328 MSI 65 28.017 CN Low 90 38.793 CN High 60 25.862 CN, copy number; MSI, microsatellite instability; POLE, DNA polymerase ε.
    [Show full text]
  • SATB2 Gene SATB Homeobox 2
    SATB2 gene SATB homeobox 2 Normal Function The SATB2 gene provides instructions for making a protein that helps control the development of certain body systems. The SATB2 protein attaches to special regions of DNA called matrix attachment regions (MARs). These regions help determine the structure of chromatin, which is the complex of DNA and proteins that packages DNA into chromosomes. The structure of chromatin is one way that gene expression is regulated during development. By organizing chromatin structure, the SATB2 protein coordinates the activity of multiple genes involved in the development of certain body systems. In particular, the SATB2 protein promotes the maturation of cells that build bones (osteoblasts) and directs development of structures in the head and face. The protein also plays roles in the maturation and function of different types of nerve cells (neurons) in the brain. Health Conditions Related to Genetic Changes SATB2-associated syndrome Mutations in the SATB2 gene have been found to cause SATB2-associated syndrome. Individuals with this condition have intellectual disability and severe speech problems. They may also have an opening in the roof of the mouth, dental abnormalities, or other abnormalities of the head and face (craniofacial anomalies). Some of these mutations are deletions of large pieces of DNA that remove several genes, including SATB2. Other mutations add, remove, or rearrange smaller pieces of DNA within the SATB2 gene. Still other mutations change single DNA building blocks (nucleotides) in the SATB2 gene. It is likely that these genetic changes reduce the amount of functional SATB2 protein. Reduction of SATB2 function is thought to impair normal development of the brain and craniofacial structures, leading to intellectual disability, delayed speech, craniofacial anomalies, and other features of SATB2-associated syndrome.
    [Show full text]
  • REVIEW Hormones and the Hippocampus
    205 REVIEW Hormones and the hippocampus R Lathe Centre for Genome Research and Centre for Neuroscience, University of Edinburgh, West Mains Road, Edinburgh EH9 3JQ, UK (Requests for offprints should be addressed to R Lathe, Centre for Genome Research, King’s Buildings, West Mains Road, Edinburgh EH9 3JQ, UK; Email: [email protected]) Abstract Hippocampal lesions produce memory deficits, but the Functionally, hippocampal enteroception may reflect feed- exact function of the hippocampus remains obscure. back control; evidence is reviewed that the hippocampus Evidence is presented that its role in memory may be modulates body physiology, including the activity of the ancillary to physiological regulation. Molecular studies hypothalamus–pituitary–adrenal axis, blood pressure, demonstrate that the hippocampus is a primary target for immunity, and reproductive function. It is suggested that ligands that reflect body physiology, including ion balance the hippocampus operates, in parallel with the amygdala, and blood pressure, immunity, pain, reproductive status, to modulate body physiology in response to cognitive satiety and stress. Hippocampal receptors are functional, stimuli. Hippocampal outputs are predominantly inhibi- probably accessible to their ligands, and mediate physio- tory on downstream neuroendocrine activity; increased logical and cognitive changes. This argues that an early synaptic efficacy in the hippocampus (e.g. long-term role of the hippocampus may have been in sensing soluble potentiation) could facilitate throughput inhibition.
    [Show full text]
  • A Network of Genetic Repression and Derepression Specifies Projection
    A network of genetic repression and derepression INAUGURAL ARTICLE specifies projection fates in the developing neocortex Karpagam Srinivasana,1, Dino P. Leonea, Rosalie K. Batesona, Gergana Dobrevab, Yoshinori Kohwic, Terumi Kohwi-Shigematsuc, Rudolf Grosschedlb, and Susan K. McConnella,2 aDepartment of Biology, Stanford University, Stanford, CA 94305; bMax-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; and cLawrence Berkeley National Laboratory, Berkeley, CA 94720 This contribution is part of the special series of Inaugural Articles by members of the National Academy of Sciences elected in 2011. Contributed by Susan K. McConnell, September 28, 2012 (sent for review August 24, 2012) Neurons within each layer in the mammalian cortex have stereotypic which results in a suppression of corticothalamic and callosal projections. Four genes—Fezf2, Ctip2, Tbr1, and Satb2—regulate fates, respectively, and axon extension along the corticospinal these projection identities. These genes also interact with each tract (CST). Fezf2 mutant neurons fail to repress Satb2 and Tbr1, other, and it is unclear how these interactions shape the final pro- thus their axons cross the CC and/or innervate the thalamus jection identity. Here we show, by generating double mutants of inappropriately. Fezf2, Ctip2, and Satb2, that cortical neurons deploy a complex In callosal projection neurons, Satb2 represses the expression Ctip2 Bhlhb5 genetic switch that uses mutual repression to produce subcortical of and , leading to a repression of subcerebral fates. Satb2 Tbr1 or callosal projections. We discovered that Tbr1, EphA4, and Unc5H3 Interestingly, we found that promotes expression in fi upper layer callosal neurons, and Tbr1 expression in these neu- are critical downstream targets of Satb2 in callosal fate speci ca- fi tion.
    [Show full text]