Transcription Factor Expression Defines Subclasses of Developing Projection Neurons Highly Similar to Single-Cell RNA-Seq Subtypes

Total Page:16

File Type:pdf, Size:1020Kb

Transcription Factor Expression Defines Subclasses of Developing Projection Neurons Highly Similar to Single-Cell RNA-Seq Subtypes Transcription factor expression defines subclasses of developing projection neurons highly similar to single-cell RNA-seq subtypes Whitney E. Heavnera,b, Shaoyi Jib, James H. Notwellc, Ethan S. Dyerd,e,f, Alex M. Tsengc, Johannes Birgmeierc, Boyoung Yooc, Gill Bejeranoc,g,h,i, and Susan K. McConnellb,1 aCenter for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101; bDepartment of Biology, Stanford University, Stanford, CA 94305; cDepartment of Computer Science, Stanford University, Stanford, CA 94305; dStanford Institute for Theoretical Physics, Stanford University, Stanford, CA 94305; eDepartment of Physics and Astronomy, The Johns Hopkins University, Baltimore, MD 21218; fGoogle, Mountain View, CA 94043; gDepartment of Developmental Biology, Stanford University, Stanford, CA 94305; hDepartment of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305; and iDepartment of Biomedical Data Science, Stanford University, Stanford, CA 94305 Contributed by Susan K. McConnell, August 12, 2020 (sent for review April 27, 2020; reviewed by Anthony Paul Barnes and Seth Blackshaw) We are only just beginning to catalog the vast diversity of cell clustering and visualization tools, such as t-distributed stochastic types in the cerebral cortex. Such categorization is a first step neighbor embedding (t-SNE) (8) and uniform manifold ap- toward understanding how diversification relates to function. All proximation and projection (UMAP) (9). Default t-SNE, how- cortical projection neurons arise from a uniform pool of progenitor ever, does not preserve global structure and therefore may miss cells that lines the ventricles of the forebrain. It is still unclear how biologically meaningful hierarchies in single-cell data (10). these progenitor cells generate the more than 50 unique types of UMAP, on the other hand, was shown to preserve more local mature cortical projection neurons defined by their distinct gene- structure and continuity between cell subtypes than t-SNE, sug- expression profiles. Moreover, exactly how and when neurons di- gesting that UMAP improves on the utility of t-SNE for yielding versify their function during development is unknown. Here we insights into cell fate trajectories (9). A single cortical progenitor relate gene expression and chromatin accessibility of two sub- cell, for example, generates many daughter cells, each of which classes of projection neurons with divergent morphological and follows a specific trajectory to become a highly specialized cell functional features as they develop in the mouse brain between type among a larger population with similar features. The cere- embryonic day 13 and postnatal day 5 in order to identify tran- bral cortex can be divided into two such large populations: scriptional networks that diversify neuron cell fate. We compare Projection neurons of the deep layers (DL), layer 6 (L6) and L5, these gene-expression profiles with published profiles of single cells isolated from similar populations and establish that layer- which are populated first, followed in order by projection neu- defined cell classes encompass cell subtypes and developmental rons of the upper layers (UL), L4, then L2/3 (11). Although DL trajectories identified using single-cell sequencing. Given the and UL subclasses are heterogeneous, cells within each subclass depth of our sequencing, we identify groups of transcription fac- share common morphological and electrophysiological proper- tors with particularly dense subclass-specific regulation and ties. The majority of DL neurons, for example, are corticofugal subclass-enriched transcription factor binding motifs. We also de- projection neurons (CFPNs), which send their axons to areas scribe transcription factor-adjacent long noncoding RNAs that de- outside the cortex, including the thalamus and the spinal cord. fine each subclass and validate the function of Myt1l in balancing The majority of UL neurons are cortico–cortical projection the ratio of the two subclasses in vitro. Our multidimensional ap- proach supports an evolving model of progressive restriction of Significance cell fate competence through inherited transcriptional identities. Neurons of the cerebral cortex arise from a common progenitor transcription | gene regulation | cortical development | pool that progressively generates different cell subtypes with next-generation sequencing distinct features, including deep layer (DL) and upper layer (UL) projection neurons. Several transcription factors (TFs) are he cerebral cortex is the region of the human brain respon- known to specify whether a progenitor cell will become UL or Tsible for perception, language, complex thinking, and motor DL; however, it is still unknown which upstream and down- control. Neurons in the cortex can be subdivided into two broad stream factors contribute to this fate decision. Here, we deeply classes: Excitatory glutamatergic projection neurons and inhibi- characterize differential gene expression and chromatin ac- tory GABAergic interneurons. Subclasses of cells exist within cessibility of UL and DL neurons. We identify TFs that are highly each of these broad classes. Recent efforts have sought to differentially expressed and densely regulated and find that UL identify the complete catalog of cell types in the cortex using neurons retain the transcriptomic signature of their mother transcriptional profiling (1–4). At least 13 unique types of ex- cells. We also discover a previously unknown role for the TF citatory neurons have been identified in the developing mouse Myt1l in cell fate specification. cortex (2), while the adult mouse cortex contains at least 52, a Author contributions: W.E.H., S.J., J.H.N., G.B., and S.K.M. designed research; W.E.H. and number that may rise with increased computational power to S.J. performed research; W.E.H., S.J., J.H.N., E.S.D., A.M.T., J.B., B.Y., G.B., and S.K.M. an- delineate cell types among single cells (1, 3). alyzed data; and W.E.H., S.J., J.H.N., E.S.D., A.M.T., J.B., G.B., and S.K.M. wrote the paper. While our ability to identify neuron subtypes contributes to Reviewers: A.P.B., Oregon Health & Science University; and S.B., The Johns Hopkins Uni- our understanding of functional divisions within cortical circuits versity School of Medicine. (5), how these circuits are specified during development remains The authors declare no competing interest. unclear. Several recent studies have used single-cell RNA se- Published under the PNAS license. quencing (scRNA-seq) to identify how differential gene expres- 1To whom correspondence may be addressed. Email: [email protected]. sion over time establishes all of the identifiable cell subtypes in This article contains supporting information online at https://www.pnas.org/lookup/suppl/ the developing brain (2, 6, 7). Delineating cell types using doi:10.1073/pnas.2008013117/-/DCSupplemental. scRNA-seq profiles relies on dimensionality reduction with First published September 18, 2020. 25074–25084 | PNAS | October 6, 2020 | vol. 117 | no. 40 www.pnas.org/cgi/doi/10.1073/pnas.2008013117 Downloaded by guest on September 26, 2021 neurons (CPNs), many of which send their axons across the midline along the corpus callosum. While CPNs overwhelmingly occupy the ULs, they can also be found in the DLs. A network of key transcription factors (TFs) expressed in early projection neurons determines whether a cell will acquire CFPN or CPN fate (12–14). Previous studies in the mouse revealed that TBR1 and FEZF2 regulate L6 and L5 CFPN development, re- spectively (15–22), while SATB2 is required for CPN fate (23–25). The upstream and downstream transcription cascade governing layer-specific maturation over time, however, is still unknown. No single study has directly assessed chromatin accessibility—an important indicator of gene regulation—and gene expression in CPNs or CFPNs during a specific stage of embryonic or early postnatal development. Moreover, it is still unknown how such transcriptional profiles differ between CPNs and CFPNs at comparable stages of development. One previous screen used bulk RNA-seq to identify genes that are differen- tially expressed between projection neuron subtypes on the same day of development in the mouse, when DL neurons are more developmentally mature than UL neurons (26). Here, in order to identify transcriptional states that determine DL versus UL fate, we chose sequencing depth over sequencing breadth and analyzed cell type-specific gene expression plus differential chromatin accessibility over three defined stages of development in two sets of genetically defined neurons. We compared subclass- and stage-defining gene sets with subtype- defining gene sets identified using scRNA-seq and found broad agreement between our bulk-sequenced neuron populations and populations identified by scRNA-seq. Given the robustness of NEUROSCIENCE bulk sequencing, we also joined open chromatin with gene ex- pression within these subclasses and identified densely regulated TFs with previously uncharacterized roles in specifying UL ver- Fig. 1. Excitatory projection neurons of the mouse neocortex can be sub- sus DL fate. In addition, we identified long-noncoding RNAs divided into two broad classes, here called upper layer (UL) and deep layer adjacent to important TF genes (TF-lncRNAs) with highly spe- (DL), each identified by a specific combination of TFs and labeled by an exclusive fluorescent reporter. (A) Schematic representation of neocortical cific spatial
Recommended publications
  • Functional Analysis of the Homeobox Gene Tur-2 During Mouse Embryogenesis
    Functional Analysis of The Homeobox Gene Tur-2 During Mouse Embryogenesis Shao Jun Tang A thesis submitted in conformity with the requirements for the Degree of Doctor of Philosophy Graduate Department of Molecular and Medical Genetics University of Toronto March, 1998 Copyright by Shao Jun Tang (1998) National Library Bibriothèque nationale du Canada Acquisitions and Acquisitions et Bibiiographic Services seMces bibliographiques 395 Wellington Street 395, rue Weifington OtbawaON K1AW OttawaON KYAON4 Canada Canada The author has granted a non- L'auteur a accordé une licence non exclusive licence alIowing the exclusive permettant à la National Library of Canada to Bibliothèque nationale du Canada de reproduce, loan, distri%uteor sell reproduire, prêter' distribuer ou copies of this thesis in microform, vendre des copies de cette thèse sous paper or electronic formats. la forme de microfiche/nlm, de reproduction sur papier ou sur format électronique. The author retains ownership of the L'auteur conserve la propriété du copyright in this thesis. Neither the droit d'auteur qui protège cette thèse. thesis nor substantial extracts fkom it Ni la thèse ni des extraits substantiels may be printed or otherwise de celle-ci ne doivent être imprimés reproduced without the author's ou autrement reproduits sans son permission. autorisation. Functional Analysis of The Homeobox Gene TLr-2 During Mouse Embryogenesis Doctor of Philosophy (1998) Shao Jun Tang Graduate Department of Moiecular and Medicd Genetics University of Toronto Abstract This thesis describes the clonhg of the TLx-2 homeobox gene, the determination of its developmental expression, the characterization of its fiuiction in mouse mesodem and penpheral nervous system (PNS) developrnent, the regulation of nx-2 expression in the early mouse embryo by BMP signalling, and the modulation of the function of nX-2 protein by the 14-3-3 signalling protein during neural development.
    [Show full text]
  • Role of Estrogen Receptor Beta and the Isoflavone Genistein
    WCP2018 OR28-3 Oral session White-to-brown adipose differentiation: role of estrogen receptor beta and the isoflavone genistein Alessandra Bitto, Federica Mannino, Natasha Irrera, Giovanni Pallio, Domenica Altavilla, Francesco Squadrito Clinical and experimental medicine, University of Messina, Italy The two types of fat cells in mammals brown and white have different functions. White adipose tissue (WAT) stores excess energy in the form of triglyceride and releases free fatty acids during caloric deficiency. Brown adipose tissue (BAT) on the other hand can dissipate energy through thermogenesis. Genistein can have an effect on energy expenditure UCP (uncoupling protein) expression and protect against the obesogenic effect of a high calorie diet. The effect of genistein in inducing white-to-brown transdifferentiation was investigated in 3T3-L1 cells differentiated into white adipocytes with a specific medium (DMEM 10% calf serum 1% penicillin/streptomycin 500 uM 3isobutyl1 methylxanthine 10ug/ml insulin 250 nM dexmethasone 8 ug/ml biotin and 4 ug/ml pantothenic acid). Fully differentiated white adipocytes were treated after 10 days with different genistein doses (10-50-100-200 uM) for 24-48h or left untreated. Two specific ER-beta and PPAR-gamma receptor inhibitors were also used to understand if genistein effects are mediated by the estrogen or the PPAR receptor. Also a CRISPR/Cas9 approach was used to delete either ER-beta or PPAR-gamma to clarify which receptor is involved in genistein action. Intracellular lipid accumulation was determined by oil-red-O staining after 24 and 48hours of treatment. The expression of UCP1 estrogen receptor alpha and beta PPARalpha and gamma DIO2 (Type II iodothyronine deiodinase) PRDM16 (PR domain containing 16) and CIDEA (cell death inducing DNA fragmentation factor) were evaluated by qPCR after 24 and 48hours of genistein treatment.
    [Show full text]
  • Hes5 Regulates the Transition Timing of Neurogenesis and Gliogenesis In
    © 2017. Published by The Company of Biologists Ltd | Development (2017) 144, 3156-3167 doi:10.1242/dev.147256 RESEARCH ARTICLE Hes5 regulates the transition timing of neurogenesis and gliogenesis in mammalian neocortical development Shama Bansod1,2, Ryoichiro Kageyama1,2,3,4 and Toshiyuki Ohtsuka1,2,3,* ABSTRACT has been reported that the high mobility group AT-hook (Hgma) During mammalian neocortical development, neural stem/progenitor genes regulate gene expression by modulating chromatin structure cells (NSCs) sequentially give rise to deep layer neurons and (Ozturk et al., 2014), maintain neurogenic NSCs, and inhibit superficial layer neurons through mid- to late-embryonic stages, gliogenesis during early- to mid-embryonic stages through global shifting to gliogenic phase at perinatal stages. Previously, we found opening of the chromatin state (Kishi et al., 2012). However, the that the Hes genes inhibit neuronal differentiation and maintain mechanism by which the expression of these epigenetic factors is NSCs. Here, we generated transgenic mice that overexpress Hes5 in controlled remains to be analyzed. NSCs of the central nervous system, and found that the transition Here, we found that Hes5, a transcriptional repressor acting timing from deep to superficial layer neurogenesis was shifted earlier, as an effector of Notch signaling, regulates the timing of while gliogenesis precociously occurred in the developing neocortex neurogenesis and gliogenesis via alteration in the expression of Hes5-overexpressing mice. By contrast, the transition from deep to levels of epigenetic factors. Notch signaling contributes to the superficial layer neurogenesis and the onset of gliogenesis were elaboration of cellular diversity during the development of various delayed in Hes5 knockout (KO) mice.
    [Show full text]
  • Ptf1a/Rbpj Complex Inhibits Ganglion Cell Fate and Drives the Specification of All Horizontal Cell Subtypes in the Chick Retina
    Ptf1a/Rbpj complex inhibits ganglion cell fate and drives the specification of all horizontal cell subtypes in the chick retina. Elise Lelièvre, Monkol Lek, Henrik Boije, L. Houille-Vernes, Valérie Brajeul, A. Slembrouck, Jérôme Roger, José-Alain Sahel, Jean-Marc Matter, Florian Sennlaub, et al. To cite this version: Elise Lelièvre, Monkol Lek, Henrik Boije, L. Houille-Vernes, Valérie Brajeul, et al.. Ptf1a/Rbpj complex inhibits ganglion cell fate and drives the specification of all horizontal cell subtypes in the chick retina.: Ptf1a in chick retinal development. Developmental Biology, Elsevier, 2011, 358 (2), pp.296-308. 10.1016/j.ydbio.2011.07.033. inserm-00614775 HAL Id: inserm-00614775 https://www.hal.inserm.fr/inserm-00614775 Submitted on 16 Aug 2011 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Ptf1a/Rbpj complex inhibits ganglion cell fate and drives the specification of all horizontal cell subtypes in the chick retina. 1,2,3,4,5 6 6 2,4,5 2,4,5 E.C. Lelièvre , M. Lek , H. Boije , L. Houille-Verne s , V. Brajeul , A. Slembrouck2,4,5, J.E. Roger4, J. Sahel2,4,5, J.M.
    [Show full text]
  • An Interactive Web Application to Explore Regeneration-Associated Gene Expression and Chromatin Accessibility
    Supplementary Materials Regeneration Rosetta: An interactive web application to explore regeneration-associated gene expression and chromatin accessibility Andrea Rau, Sumona P. Dhara, Ava J. Udvadia, Paul L. Auer 1. Table S1. List of cholesterol metabolic genes from MGI database 2. Table S2. List of differentially expressed transcripts during optic nerve regeneration in zebrafish using the MGI cholesterol metabolic gene queries in the Regeneration Rosetta app 3. Table S3. List of transcription factor encoding genes from brain cell bodies following spinal cord injury in lamprey over a course of 12 weeKs 4. Table S4. List of transcription factor encoding genes from spinal cell bodies following spinal cord injury in lamprey over a course of 12 weeks Ensembl ID MGI Gene ID Symbol Name ENSMUSG00000015243 MGI:99607 Abca1 ATP-binding cassette, sub-family A (ABC1), member 1 ENSMUSG00000026944 MGI:99606 Abca2 ATP-binding cassette, sub-family A (ABC1), member 2 ENSMUSG00000024030 MGI:107704 Abcg1 ATP binding cassette subfamily G member 1 ENSMUSG00000026003 MGI:87866 Acadl acyl-Coenzyme A dehydrogenase, long-chain ENSMUSG00000018574 MGI:895149 Acadvl acyl-Coenzyme A dehydrogenase, very long chain ENSMUSG00000038641 MGI:2384785 Akr1d1 aldo-keto reductase family 1, member D1 ENSMUSG00000028553 MGI:1353627 Angptl3 angiopoietin-like 3 ENSMUSG00000031996 MGI:88047 Aplp2 amyloid beta (A4) precursor-like protein 2 ENSMUSG00000032083 MGI:88049 Apoa1 apolipoprotein A-I ENSMUSG00000005681 MGI:88050 Apoa2 apolipoprotein A-II ENSMUSG00000032080 MGI:88051 Apoa4
    [Show full text]
  • Letters to the Editor
    LETTERS TO THE EDITOR The closely related rare and severe acute myeloid leukemias carrying EVI1 or PRDM16 rearrangements share singular biological features In a recent issue of Haematologica , Matsuo et al .1 pinpoint the pejorative effect of EVI1 overexpression in 18 acute myeloid leukemias (AML) with MLL rearrangements. However, EVI1 overexpression has also been reported in patients with translocations involving chromosome 3 and the EVI1 gene. 2,3 Because of the poor prognosis associated to these anomalies, it is important to investigate them at an early stage in order to adapt patient management. Indeed, previous reports 4-6 and the 2008 WHO classification 7 indi - cate that EVI1-rearranged (EVI1-r) AML display typical fea - tures, such as absence of thrombopenia, atypical megakary - Figure 1. Algorithm for the suspicion of EVI1 and PRDM16 AMLs. ocytes and multilineage dysplasia 2-4 which can be detected by current diagnostic reference methods. In this line, we compared a cohort of 17 EVI1-r AML, aged between 8 and 79-years old (median 54 years) to 1822 other cases of AML months. diagnosed in the same laboratory over 14 years. At diagno - This study consolidates the unusual base-line character - sis, there were similar hemoglobin levels or white blood istics and clinical features of EVI1-r AML cases. Moreover, cell counts in both groups. Median platelet counts were it indicates a very low rate of MPO expression in EVI1-r 9 9 123x10 /L, higher than 100x10 /L in 53% of EVI1-r AML AML patients. It is interesting to note that relationships patients, compared to 25% in the control AML population have been reported between EVI1 expression and MPO (P=0.02).
    [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]
  • ARID5B As a Critical Downstream Target of the TAL1 Complex That Activates the Oncogenic Transcriptional Program and Promotes T-Cell Leukemogenesis
    Downloaded from genesdev.cshlp.org on October 10, 2021 - Published by Cold Spring Harbor Laboratory Press ARID5B as a critical downstream target of the TAL1 complex that activates the oncogenic transcriptional program and promotes T-cell leukemogenesis Wei Zhong Leong,1 Shi Hao Tan,1 Phuong Cao Thi Ngoc,1 Stella Amanda,1 Alice Wei Yee Yam,1 Wei-Siang Liau,1 Zhiyuan Gong,2 Lee N. Lawton,1 Daniel G. Tenen,1,3,4 and Takaomi Sanda1,4 1Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore; 2Department of Biological Sciences, National University of Singapore, 117543 Singapore; 3Harvard Medical School, Boston, Massachusetts 02215, USA; 4Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore The oncogenic transcription factor TAL1/SCL induces an aberrant transcriptional program in T-cell acute lym- phoblastic leukemia (T-ALL) cells. However, the critical factors that are directly activated by TAL1 and contribute to T-ALL pathogenesis are largely unknown. Here, we identified AT-rich interactive domain 5B (ARID5B) as a col- laborating oncogenic factor involved in the transcriptional program in T-ALL. ARID5B expression is down-regulated at the double-negative 2–4 stages in normal thymocytes, while it is induced by the TAL1 complex in human T-ALL cells. The enhancer located 135 kb upstream of the ARID5B gene locus is activated under a superenhancer in T-ALL cells but not in normal T cells. Notably, ARID5B-bound regions are associated predominantly with active tran- scription. ARID5B and TAL1 frequently co-occupy target genes and coordinately control their expression.
    [Show full text]
  • Id4: an Inhibitory Function in the Control
    Id4: an inhibitory function in the control of hair cell formation? Sara Johanna Margarete Weber Thesis submitted to the University College London (UCL) for the degree of Master of Philosophy The work presented in this thesis was conducted at the UCL Ear Institute between September 23rd 2013 and October 19th 2015. 1st Supervisor: Dr Nicolas Daudet UCL Ear Institute, London, UK 2nd Supervisor: Dr Stephen Price UCL Department of Cell and Developmental Biology, London, UK 3rd Supervisor: Prof Guy Richardson School of Life Sciences, University of Sussex, Brighton, UK I, Sara Weber confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Heidelberg, 08.03.2016 ………………………….. Sara Weber 2 Abstract Mechanosensitive hair cells in the sensory epithelia of the vertebrate inner ear are essential for hearing and the sense of balance. Initially formed during embryological development they are constantly replaced in the adult avian inner ear after hair cell damage and loss, while practically no spontaneous regeneration occurs in mammals. The detailed molecular mechanisms that regulate hair cell formation remain elusive despite the identification of a number of signalling pathways and transcription factors involved in this process. In this study I investigated the role of Inhibitor of differentiation 4 (Id4), a member of the inhibitory class V of bHLH transcription factors, in hair cell formation. I found that Id4 is expressed in both hair cells and supporting cells of the chicken and the mouse inner ear at stages that are crucial for hair cell formation.
    [Show full text]
  • Epigenetic Mechanisms of Lncrnas Binding to Protein in Carcinogenesis
    cancers Review Epigenetic Mechanisms of LncRNAs Binding to Protein in Carcinogenesis Tae-Jin Shin, Kang-Hoon Lee and Je-Yoel Cho * Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; [email protected] (T.-J.S.); [email protected] (K.-H.L.) * Correspondence: [email protected]; Tel.: +82-02-800-1268 Received: 21 September 2020; Accepted: 9 October 2020; Published: 11 October 2020 Simple Summary: The functional analysis of lncRNA, which has recently been investigated in various fields of biological research, is critical to understanding the delicate control of cells and the occurrence of diseases. The interaction between proteins and lncRNA, which has been found to be a major mechanism, has been reported to play an important role in cancer development and progress. This review thus organized the lncRNAs and related proteins involved in the cancer process, from carcinogenesis to metastasis and resistance to chemotherapy, to better understand cancer and to further develop new treatments for it. This will provide a new perspective on clinical cancer diagnosis, prognosis, and treatment. Abstract: Epigenetic dysregulation is an important feature for cancer initiation and progression. Long non-coding RNAs (lncRNAs) are transcripts that stably present as RNA forms with no translated protein and have lengths larger than 200 nucleotides. LncRNA can epigenetically regulate either oncogenes or tumor suppressor genes. Nowadays, the combined research of lncRNA plus protein analysis is gaining more attention. LncRNA controls gene expression directly by binding to transcription factors of target genes and indirectly by complexing with other proteins to bind to target proteins and cause protein degradation, reduced protein stability, or interference with the binding of other proteins.
    [Show full text]
  • The Effect of Chromosomal Translocations in Acute Leukemias: the LMO2 Paradigm in Transcription and Development I
    [CANCER RESEARCH (SUPPL.) 59, 1794s-1798s, April 1, 1999] The Effect of Chromosomal Translocations in Acute Leukemias: The LMO2 Paradigm in Transcription and Development I Terence H. Rabbitts, 2 Katharina Bucher, Grace Chung, Gerald Grutz, Alan Warren, and Yoshi Yamada Medical Research Council Laboratory of Molecular Biology, Division of Protein and Nucleic Acid Chemistry, Hills Road, Cambridge, CB2 2QH United Kingdom Abstract proto-oncogene from a Burkitt's lymphoma translocation breakpoint t(8;14)(q24;q32) revealed that, while the translocated gene was intact, Two general features have emerged about genes that are activated after there were many mutations in both the noncoding first exon and chromosomal translocations in acute forms of cancer. The protein prod- within the coding region itself (4-11). In T-cell acute leukemias, there ucts of these genes are transcription regulators and are involved in are several different chromosomal translocations found in individual developmental processes, and it seems that the subversion of these normal functions accounts for their role in tumorigenesis. The features of the tumors, and the analysis of these has led to the discovery of many LMO family of genes, which encode LIM-domain proteins involved in different, novel genes that can contribute to T-cell tumorigenesis (1). T-cell acute leukemia through chromosomal translocations, typify these These and many other studies have led to two conclusions: abnormal functions in tumorigenesis. For example, the LMO2 protein is (a) many of the chromosomal translocation-activated oncogenes are involved in the formation of multimeric DNA-binding complexes, which transcription regulators in their normal sites of expression, and it is may vary in composition at different stages of hematopoiesis and function this property that is instrumental in their involvement in tumor etiol- to control differentiation of specific lineages.
    [Show full text]
  • Modulation of the Activity of a Key Metabolic Regulator Small Heterodimer Partner by Post-Translational Modifications
    MODULATION OF THE ACTIVITY OF A KEY METABOLIC REGULATOR SMALL HETERODIMER PARTNER BY POST-TRANSLATIONAL MODIFICATIONS BY DEEPTHI KANAMALURU DISSERTATION Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biochemistry in the Graduate College of the University of Illinois at Urbana-Champaign, 2011 Urbana, Illinois Doctoral Committee: Associate Professor Jongsook Kim Kemper, Chair Professor David J. Shapiro Professor Milan K. Bagchi Assistant Professor Lin-Feng Chen Abstract Small Heterodimer Partner (SHP, NR0B2), a member of the nuclear receptor superfamily, is an orphan receptor that lacks a DNA binding domain but contains a putative ligand binding domain. SHP forms non-functional heterodimers with DNA binding transcriptional factors and, thereby, functions as a transcriptional corepressor in diverse biological processes, including cellular metabolism, cell proliferation, apoptosis, and sexual maturation. Of these reported functions of SHP, maintaining cholesterol and bile acid levels by negative feedback regulation of hepatic conversion of cholesterol to bile acids is well established. Cholesterol is essential in many biological activities in mammalian cells. Conversion of hepatic cholesterol into bile acids is a major pathway to eliminate cholesterol from the body. However, excess amounts of cholesterol and bile acids are pathogenic. Therefore, the levels of cholesterol and bile acids need to be tightly regulated. Cholesterol 7α-hydroxylase (CYP7A1), a liver specific P450 enzyme, is the first and rate-limiting enzyme in this process. Increased levels of bile acids repress transcription of CYP7A1 in a feedback manner. In response to elevated bile acid levels, the nuclear bile acid receptor Farnesoid X Receptor (FXR) increases the transcription of SHP.
    [Show full text]