Snapshot: Ca2+-Dependent Transcription in Neurons Janine Zieg, Paul L

Total Page:16

File Type:pdf, Size:1020Kb

Snapshot: Ca2+-Dependent Transcription in Neurons Janine Zieg, Paul L SnapShot: Ca2+-Dependent Transcription in Neurons Janine Zieg, Paul L. Greer, and Michael E. Greenberg Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA See online version for legend and references. Also see the September 25, 2008, Neuron Reviews Issue 1080 Cell 134, September 19, 2008 ©2008 Elsevier Inc. DOI 10.1016/j.cell.2008.09.010 entitled “Calcium Signaling at the Synapse.” SnapShot: Ca2+-Dependent Transcription in Neurons Janine Zieg, Paul L. Greer, and Michael E. Greenberg Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA Synaptic activity stimulates the influx of calcium ions into the postsynaptic neuron and thereby sets in motion a cascade of signaling events that lead to changes in gene expression. These changes in gene expression affect many aspects of nervous system development including dendritic morphogenesis, neuronal survival, and synapse development as well as the adaptive responses that underlie learning and memory in the mature nervous system. Mutations in components of the signal- ing pathways that participate in the process of experience-dependent brain development have been found to give rise to a variety of disorders of cognitive function including autism spectrum disorders. The initial contact between the axon and the dendrite during synapse development is mediated by cell surface-associated proteins on the pre- and postsynaptic membranes. For example, the binding of presynaptic β-neurexin to its postsynaptic receptor, Neuroligin1, leads to the recruitment of PSD-95 at nascent excitatory synapses. Ephrin/Eph signaling leads to the recruitment of additional proteins and the potentiation of NMDA receptor signaling. Release of the excitatory neurotrans- mitter glutamate from the presynaptic membrane and its binding to NMDA receptors and AMPA receptors on the postsynaptic membrane lead to the opening of these glutamate-gated ion channels. This is followed by membrane depolarization, opening of the L-type voltage-gated calcium ion channel (L-VSCC), and a rapid rise in calcium ions in the postsynaptic neuron as well as other local changes including protein recruitment and activation. Calcium entry through L-VSCCs leads to the recruitment of AKAP79/150, which then recruits PKA to the channel. PKA phosphorylates the calcium channel, thereby increasing its ability to allow calcium ions to flow into the cell. Calcium ion influx through L-VSCCs is sensed by calmodulin (CaM). Activated calmodulin initiates a cascade of events including stimulation of the guanine nucleotide exchange factor RasGRF, followed by activation of the Ras-MAPK signaling cascade. Calcium-activated calmodulin also activates the CaM kinase signaling pathway. Once activated these pathways trigger the phosphorylation and activation of a wide range of transcription factors such as CREB and NeuroD2. The phosphorylation of these transcription factors occurs in the nucleus and can be triggered by a cascade of events that begins at the site of calcium entry at the mouth of the calcium channel (that is, the Ras, Raf, MEK, ERK, RSK/MSK signaling pathway). Alternatively, channel activation can trigger an elevation of calcium ions directly in the nucleus that leads to activation of nuclear CaMKII by calcium/calmodulin, which in turn phosphorylates CREB and NeuroD2. In addition, dephosphorylation- dependent signaling through calcium/calmodulin activation of calcineurin leads to the activation of the transcripton factors NFAT and MEF2. Once activated and localized to the nucleus, calcium-activated transcription factors and modulators of transcription bind to the regulatory regions of activity- regulated genes to orchestrate finely tuned levels of gene expression. The most extensively studied activity-regulated gene isBdnf , the gene encoding brain-derived neurotrophic factor. BDNF affects numerous processes in neuronal development including axonal and dendritic development, synapse formation and maturation, syn- aptic potentiation, and neuronal survival. Polymorphisms in the Bdnf gene correlate with defects in learning and memory. The promoter region of Bdnf is complex and includes at least seven different 5′ exons, only some of which are regulated by synaptic activity. At activity-regulated promoter IV, CREB, USF1/2, and CaRF occupy three distinct calcium response elements: CaRE1, CaRE2, and CaRE3. Neuronal activity leads to the activation of CREB by phosphorylation at three serine residues (Ser133, 142, and 143) through the combined action of kinases including RSK, CaMKII, and CaMKIV. Activated CREB binds to its cofactor, the histone acetyl trans- ferase CBP, to promote Bdnf transcription. CREB activation of Bdnf transcription may be modulated by CREM, which is itself an activity-regulated CREB-dependent gene. MEF2, activated through dephosphoryation by calcineurin, positively regulates Bdnf expression. Prior to neurotransmitter release onto the postsynaptic neuron, MEF2 binds to transcriptional corepressors mSin3a, SUV39H, and HDACs. NF-κB, activated by CaMKII-dependent phosphorylation in the cytoplasm, translocates to the nucleus to positively regulate Bdnf expression. Npas4, a PAS-domain bHLH transcription factor known to promote inhibitory synapse formation on excitatory neurons, also binds to Bdnf promoter IV and appears to sustain BDNF expression. Translation of MeCP2 mRNA is regulated by a CREB-dependent microRNA, and the MeCP2 protein, a methyl-CpG-binding protein, is phosphorylated by CaMKII at Ser421. MeCP2 modulation of Bdnf expression is complex and is important in light of the involvement of MeCP2 in the etiology of Rett Syndrome. The complex of proteins that regulates Bdnf expression changes upon the influx of calcium ions into the postsynaptic neuron, in part through the CaMK-dependent phosphorylation of HDACs that control chromatin organization in the vicinity of Bdnf promoter IV. The release of HDACs is accompanied by the recruitment of the CREB-binding protein CBP, a histone acetyltransferase that engages the polymerase II transcriptional machinery. In addition to Bdnf, the expression of hundreds of other genes is also regulated by synaptic activity. Although the function of some of these genes is not understood, it is known that the activity-dependent regulation of gene expression promotes many diverse processes such as neuronal survival, dendrite formation, synaptic devel- opment, and adaptive responses. Interestingly, many of the molecules involved in these activity-dependent signal transduction pathways leading to activity-regulated gene expression are now known to be mutated in diseases of cognition such as autism spectrum disorders, suggesting that this activity-dependent program is im- portant for human nervous system development. Abbreviations AKAP, A-kinase anchor proteins; AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; Arc, activity-regulated cytoskeleton-associated pro- tein; Bcl, cell survival protein, name derived from B cell lymphoma 2; BDNF, brain-derived neurotrophic factor; bHLHB2, basic helix-loop-helix B2 transcription factor; CaM, calmodulin; CaMK, Calcium/calmodulin-dependent protein kinase; CaMKK, calcium/calmodulin-dependent protein kinase kinase; CaRF, calcium-responsive transcription factor; CBP, CREB-binding protein; c-fos, an immediate early gene transcription factor; CREB, cyclic AMP response element-binding protein; CREM, cyclic AMP response element modulator; DREAM, downstream regulatory element-antagonist modulator; ERK, extracellular signal-regulated kinases; HDAC, his- tone deacetylase; IAP, inhibitor of apoptosis; Kalirin-7, a Rho guanine nucleotide exchange factor that interacts with huntingtin-associated protein; L-VSCC, L-type voltage-sensitive calcium channel; MeCP2, methyl-CpG-binding protein 2; MEF2, myocyte enhancer factor 2; MEK, mitogen-activated protein kinase kinase; MiR132, microRNA132; MnSOD, manganese superoxide dismutase; mSin3a, mouse Sin3a transcription regulator; NeuroD2, neurogenic differentiation 2; NFAT, nuclear factor of activated T cells; NF-kB, nuclear factor kappa B; NMDAR, N-methyl-D-aspartic acid receptor; Npas4, neuronal PAS-domain bHLH transcription factor; Nur77, nerve growth factor IB; Pdyn, prodynorphin; Rac, member of the Rho family of GTPases; Raf, ras-activated factor; Ras, ras proto-oncogene; RasGRF, ras protein- specific guanine nucleotide-releasing factor; RSK, ribosomal s6 kinase; Src, proto-oncogenic tyrosine kinase similar to v-Src protein from Rous Sarcoma Virus; Tiam, T cell lymphoma invasion and metastasis-inducing protein; USF, upstream stimulatory factor; Wnt2, wingless-type MMTV integration site family, member 2; ?, as- sociations have been shown in nonneuronal cell types. REFERENCES Chahrour, M., Jung, S.Y., Shaw, C., Zhou, X., Wong, S.T., Qin, J., and Zoghbi, H.Y. (2008). MeCP2, a key contributor to neurological disease, activates and represses transcription. Science 320, 1224–1229. Flavell, S.W., Cowan, C.W., Kim, T.K., Greer, P.L., Lin, Y., Paradis, S., Griffith, E.C., Hu, L.S., Chen, C., and Greenberg, M.E. (2006). Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science 311, 1008–1012. Husi, H., Ward, M.A., Choudhary, J.S., Blackstock, W.P., and Grant, S.G. (2000). Proteomic analysis of NMDA receptor-adhesion protein signaling complexes. Nat. Neurosci. 3, 661–669. Impey, S., McCorkle, S.R., Cha-Molstad, H., Dwyer, J.M., Yochum, G.S., Boss, J.M., McWeeney, S., Dunn, J.J., Mandel, G., and Goodman, R.H. (2004). Defining the
Recommended publications
  • SF3B3) and Sin3a Associated Protein 130 (SAP130
    cells Communication Ambiguity about Splicing Factor 3b Subunit 3 (SF3B3) and Sin3A Associated Protein 130 (SAP130) Paula I. Metselaar 1,* , Celine Hos 1, Olaf Welting 1, Jos A. Bosch 2,3, Aletta D. Kraneveld 4 , Wouter J. de Jonge 1 and Anje A. Te Velde 1 1 Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam UMC, University of Amsterdam, 1105BK Amsterdam, The Netherlands; [email protected] (C.H.); [email protected] (O.W.); [email protected] (W.J.d.J.); [email protected] (A.A.T.V.) 2 Department of Psychology, University of Amsterdam, 1018WS Amsterdam, The Netherlands; [email protected] 3 Department of Medical Psychology, Amsterdam UMC, University of Amsterdam, 1001NK Amsterdam, The Netherlands 4 Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; [email protected] * Correspondence: [email protected] Abstract: In 2020, three articles were published on a protein that can activate the immune system by binding to macrophage-inducible C-type lectin receptor (Mincle). In the articles, the protein was referred to as ‘SAP130, a subunit of the histone deacetylase complex.’ However, the Mincle ligand the authors aimed to investigate is splicing factor 3b subunit 3 (SF3B3). This splicing factor is unrelated to SAP130 (Sin3A associated protein 130, a subunit of the histone deacetylase-dependent Sin3A corepressor complex). The conclusions in the three articles were formulated for SF3B3, Citation: Metselaar, P.I.; Hos, C.; while the researchers used qPCR primers and antibodies against SAP130.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. 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 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • Loss of Fam60a, a Sin3a Subunit, Results in Embryonic Lethality and Is Associated with Aberrant Methylation at a Subset of Gene
    RESEARCH ARTICLE Loss of Fam60a, a Sin3a subunit, results in embryonic lethality and is associated with aberrant methylation at a subset of gene promoters Ryo Nabeshima1,2, Osamu Nishimura3,4, Takako Maeda1, Natsumi Shimizu2, Takahiro Ide2, Kenta Yashiro1†, Yasuo Sakai1, Chikara Meno1, Mitsutaka Kadota3,4, Hidetaka Shiratori1†, Shigehiro Kuraku3,4*, Hiroshi Hamada1,2* 1Developmental Genetics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; 2Laboratory for Organismal Patterning, RIKEN Center for Developmental Biology, Kobe, Japan; 3Phyloinformatics Unit, RIKEN Center for Life Science Technologies, Kobe, Japan; 4Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan Abstract We have examined the role of Fam60a, a gene highly expressed in embryonic stem cells, in mouse development. Fam60a interacts with components of the Sin3a-Hdac transcriptional corepressor complex, and most Fam60a–/– embryos manifest hypoplasia of visceral organs and die in utero. Fam60a is recruited to the promoter regions of a subset of genes, with the expression of these genes being either up- or down-regulated in Fam60a–/– embryos. The DNA methylation level of the Fam60a target gene Adhfe1 is maintained at embryonic day (E) 7.5 but markedly reduced at –/– *For correspondence: E9.5 in Fam60a embryos, suggesting that DNA demethylation is enhanced in the mutant. [email protected] (SK); Examination of genome-wide DNA methylation identified several differentially methylated regions, [email protected] (HH) which were preferentially hypomethylated, in Fam60a–/– embryos. Our data suggest that Fam60a is †These authors contributed required for proper embryogenesis, at least in part as a result of its regulation of DNA methylation equally to this work at specific gene promoters.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • A Role for Mammalian Sin3 in Permanent Gene Silencing
    Molecular Cell Article A Role for Mammalian Sin3 in Permanent Gene Silencing Chris van Oevelen,1 Jinhua Wang,1 Patrik Asp,1 Qin Yan,2,3 William G. Kaelin, Jr.,2,3 Yuval Kluger,1,* and Brian David Dynlacht1,* 1New York University School of Medicine, NYU Cancer Institute, 522 1st Avenue, New York, NY 10016, USA 2Howard Hughes Medical Institute 3Department of Medical Oncology Dana Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA *Correspondence: [email protected] (B.D.D.), [email protected] (Y.K.) DOI 10.1016/j.molcel.2008.10.015 SUMMARY substoichiometric regulatory proteins, including Swi/Snf-remod- eling proteins, retinoblastoma (RB)-binding protein 2 (RBP2), and The multisubunit Sin3 corepressor complex regu- other proteins (Hayakawa et al., 2007; Nagl et al., 2007; Sif et al., lates gene transcription through deacetylation of nu- 2001). Interestingly, RBP2 was recently shown to be a demethy- cleosomes. However, the full range of Sin3 activities lase specific for di- and trimethylated lysine 4 of histone H3 and targets is not well understood. Here, we have (Christensen et al., 2007; Klose et al., 2007). Thus, the Sin3 investigated genome-wide binding of mouse Sin3 complex provides a versatile platform for chromatin modifying and RBP2 as well as histone modifications and nucle- and remodeling activities. Sin3/Rpd3 corepressor complexes are recruited to promoter osome positioning as a function of myogenic differ- regions via sequence-specific repressors such as Ume6 or entiation. Remarkably, we find that Sin3 complexes Mad in yeast and mammalian cells, respectively, resulting in spread immediately downstream of the transcription localized deacetylation of histones within promoter regions and start site on repressed and transcribed genes during transcriptional silencing (Ayer et al., 1995; Kadosh and Struhl, differentiation.
    [Show full text]
  • CRL4B Interacts with and Coordinates the SIN3A-HDAC Complex To
    ß 2014. Published by The Company of Biologists Ltd | Journal of Cell Science (2014) 127, 4679–4691 doi:10.1242/jcs.154245 RESEARCH ARTICLE CRL4B interacts with and coordinates the SIN3A-HDAC complex to repress CDKN1A and drive cell cycle progression Qinghong Ji, Huili Hu, Fan Yang, Jupeng Yuan, Yang Yang, Liangqian Jiang, Yanyan Qian, Baichun Jiang, Yongxin Zou, Yan Wang, Changshun Shao and Yaoqin Gong* ABSTRACT Shahbazian and Grunstein, 2007). HATs catalyze the acetylation of histones and other proteins, whereas HDACs catalyze the CUL4B, a scaffold protein that assembles the CRL4B ubiquitin removal of the acetyl moieties from acetylated proteins. To date, ligase complex, participates in the regulation of a broad spectrum of 18 mammalian HDAC isoforms have been characterized and are biological processes. Here, we demonstrate a crucial role of CUL4B classified into class I, class II, class III and class IV (de Ruijter in driving cell cycle progression. We show that loss of CUL4B et al., 2003). Among them, HDAC1 and HDAC2, members of results in a significant reduction in cell proliferation and causes G1 class I, represent two of the best-characterized HDACs to date. cell cycle arrest, accompanied by the upregulation of the cyclin- They function in a number of deacetylase complexes – including dependent kinase (CDK) inhibitors (CKIs) p21 and p57 (encoded by SIN3A-HDAC, NuRD-HDAC, the BCH10-containing complex CDKN1A and CDKN1C, respectively). Strikingly, CUL4B was found and the CoREST-HDAC complex – and they are generally to negatively regulate the function of p21 through transcriptional associated with transcriptional repression (Hayakawa and repression, but not through proteolysis.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Chromatin-Associated Protein SIN3B Prevents Prostate Cancer Progression by Inducing Senescence Anthony J
    Published OnlineFirst August 14, 2017; DOI: 10.1158/0008-5472.CAN-16-3410 Cancer Tumor and Stem Cell Biology Research Chromatin-Associated Protein SIN3B Prevents Prostate Cancer Progression by Inducing Senescence Anthony J. Bainor1, Fang-Ming Deng2, Yu Wang1, Peng Lee2,4, David J. Cantor1, Susan K. Logan1,3,4, and Gregory David1,3,4 Abstract Distinguishing between indolent and aggressive prostate ade- cinoma. Furthermore, SIN3B was downregulated in human pros- nocarcinoma remains a priority to accurately identify patients tate adenocarcinoma correlating with upregulation of its target who need therapeutic intervention. SIN3B has been implicated in genes. Our results suggest a tumor suppressor function for the initiation of senescence in vitro. Here we show that in a mouse SIN3B that limits prostate adenocarcinoma progression, with model of prostate cancer, SIN3B provides a barrier to malignant potential implications for the use of SIN3B and its target genes progression. SIN3B was required for PTEN-induced cellular senes- as candidate diagnostic markers to distinguish indolent from cence and prevented progression to invasive prostate adenocar- aggressive disease. Cancer Res; 77(19); 1–10. Ó2017 AACR. Introduction damage, activation of oncogenes, or loss of a tumor suppres- sor (5, 6). Senescent cells have been identified in preneoplastic Prostate adenocarcinoma is the second most common cancer lesions of several solid tumor types, including prostatic intrae- type in American men with approximately 230,000 new pithelial neoplasias (PIN), but are rarely found in normal patients diagnosed each year, equating to about 1 in 7 men prostate or prostate adenocarcinoma (7). On the basis of these being diagnosed with prostate adenocarcinoma in his lifetime findings, cellular senescence has been hypothesized to prevent (1).
    [Show full text]
  • Transcriptional Corepressor SIN3A Regulates Hippocampal Synaptic Plasticity Via Homer1/Mglur5 Signaling
    Transcriptional corepressor SIN3A regulates hippocampal synaptic plasticity via Homer1/mGluR5 signaling Morgan Bridi, … , Nelson Spruston, Ted Abel JCI Insight. 2020;5(5):e92385. https://doi.org/10.1172/jci.insight.92385. Research Article Genetics Neuroscience Graphical abstract Find the latest version: https://jci.me/92385/pdf RESEARCH ARTICLE Transcriptional corepressor SIN3A regulates hippocampal synaptic plasticity via Homer1/mGluR5 signaling Morgan Bridi,1 Hannah Schoch,2 Cédrick Florian,3 Shane G. Poplawski,4 Anamika Banerjee,5 Joshua D. Hawk,1 Giulia S. Porcari,3 Camille Lejards,6 Chang-Gyu Hahn,5 Karl-Peter Giese,7 Robbert Havekes,3 Nelson Spruston,8 and Ted Abel3 1Mahoney Institute for Neurosciences, 2Cell and Molecular Biology Graduate Group, 3Department of Biology, 4Pharmacology Graduate Group, and 5Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA. 6Université Paul Sabatier, Toulouse, France. 7King’s College, London, United Kingdom. 8Howard Hughes Medical Institute (HHMI) Janelia Research Campus, Ashburn, Virginia, USA. Long-term memory depends on the control of activity-dependent neuronal gene expression, which is regulated by epigenetic modifications. The epigenetic modification of histones is orchestrated by the opposing activities of 2 classes of regulatory complexes: permissive coactivators and silencing corepressors. Much work has focused on coactivator complexes, but little is known about the corepressor complexes that suppress the expression of plasticity-related genes. Here, we define a critical role for the corepressor SIN3A in memory and synaptic plasticity, showing that postnatal neuronal deletion of Sin3a enhances hippocampal long-term potentiation and long- term contextual fear memory. SIN3A regulates the expression of genes encoding proteins in the postsynaptic density.
    [Show full text]
  • Tissue-Specific Metabolic Regulation of FOXO-Binding Protein
    cells Review Tissue-Specific Metabolic Regulation of FOXO-Binding Protein: FOXO Does Not Act Alone Noriko Kodani 1 and Jun Nakae 2,* 1 Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; [email protected] 2 Department of Physiology, International University of Health and Welfare School of Medicine, Narita 286-8686, Japan * Correspondence: [email protected]; Tel.: +81-476-20-7701 Received: 17 January 2020; Accepted: 10 March 2020; Published: 13 March 2020 Abstract: The transcription factor forkhead box (FOXO) controls important biological responses, including proliferation, apoptosis, differentiation, metabolism, and oxidative stress resistance. The transcriptional activity of FOXO is tightly regulated in a variety of cellular processes. FOXO can convert the external stimuli of insulin, growth factors, nutrients, cytokines, and oxidative stress into cell-specific biological responses by regulating the transcriptional activity of target genes. However, how a single transcription factor regulates a large set of target genes in various tissues in response to a variety of external stimuli remains to be clarified. Evidence indicates that FOXO-binding proteins synergistically function to achieve tightly controlled processes. Here, we review the elaborate mechanism of FOXO-binding proteins, focusing on adipogenesis, glucose homeostasis, and other metabolic regulations in order to deepen our understanding and to identify a novel therapeutic target for the prevention and treatment of metabolic disorders. Keywords: FOXO; transcription factor; FOXO-binding protein 1. Introduction Forkhead box (FOXO) transcription factors play important roles in apoptosis, the cell cycle, DNA damage repair, oxidative stress, cell differentiation, glucose metabolism, and other cellular functions [1].
    [Show full text]
  • Genome-Wide Analyses Identify Transcription Factors Required for Proper Morphogenesis of Drosophila Sensory Neuron Dendrites
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press Genome-wide analyses identify transcription factors required for proper morphogenesis of Drosophila sensory neuron dendrites Jay Z. Parrish,1 Michael D. Kim,1 Lily Yeh Jan, and Yuh Nung Jan2 Departments of Physiology and Biochemistry, Howard Hughes Medical Institute, University of California, San Francisco, California 94143, USA Dendrite arborization patterns are critical determinants of neuronal function. To explore the basis of transcriptional regulation in dendrite pattern formation, we used RNA interference (RNAi) to screen 730 transcriptional regulators and identified 78 genes involved in patterning the stereotyped dendritic arbors of class I da neurons in Drosophila. Most of these transcriptional regulators affect dendrite morphology without altering the number of class I dendrite arborization (da) neurons and fall primarily into three groups. Group A genes control both primary dendrite extension and lateral branching, hence the overall dendritic field. Nineteen genes within group A act to increase arborization, whereas 20 other genes restrict dendritic coverage. Group B genes appear to balance dendritic outgrowth and branching. Nineteen group B genes function to promote branching rather than outgrowth, and two others have the opposite effects. Finally, 10 group C genes are critical for the routing of the dendritic arbors of individual class I da neurons. Thus, multiple genetic programs operate to calibrate dendritic coverage, to coordinate the elaboration of primary versus secondary branches, and to lay out these dendritic branches in the proper orientation. [Keywords: Transcription; RNAi; Drosophila; neuron; dendrite] Supplemental material is available at http://www.genesdev.org.
    [Show full text]
  • Hats and Hdacs in Neurodegeneration: a Tale of Disconcerted Acetylation Homeostasis
    Cell Death and Differentiation (2006) 13, 539–550 & 2006 Nature Publishing Group All rights reserved 1350-9047/06 $30.00 www.nature.com/cdd Review HATs and HDACs in neurodegeneration: a tale of disconcerted acetylation homeostasis RN Saha1 and K Pahan*,1 Introduction 1 Section of Neuroscience, Department of Oral Biology, University of Nebraska Histone acetyltransferases (HATs) and histone deacetylases Medical Center, Lincoln, NE 68583-0740, USA (HDACs) represent two enzyme classes that, respectively, * Corresponding author: K Pahan, Section of Neuroscience, Department of Oral catalyze forward and backward reaction kinetics of lysine Biology, University of Nebraska Medical Center, 40th and Holdrege, Lincoln, residue acetylation in specific protein substrates. These NE 68583-0740, USA. Tel: þ 1-402-472-1324; Fax: þ 1-402-472-2551; substrates most importantly include nucleosomal histones E-mail: [email protected] and various transcription factors (TFs), which form part of Received 28.4.05; revised 11.7.05; accepted 01.8.05; published online 28.10.05 the transcription initiation complex. Accordingly, HATs and Edited by L Greene HDACs are found embedded in large multiprotein complexes near euchromatic regions of the chromatin. HATs modify Abstract core histone tails by post-translational acetylation of specific lysine residues, thereby creating appropriate ‘histone code’ Gradual disclosure of the molecular basis of selective for chromatin modification and enhanced DNA accessibility of neuronal apoptosis during neurodegenerative diseases TFs. Moreover, TFs like RelA, E2F, p53 and GATA1 are also reveals active participation of acetylating and deacetylating acetylated by HATs. In addition to enhancing protein stability, agents during the process. Several studies have now such acetylation, in most cases, enhance their transactivation successfully manipulated neuronal vulnerability by influen- potential by facilitating their interactions with DNA and other cing the dose and enzymatic activity of histone acetyltrans- proteins of the transcription apparatus.
    [Show full text]