51
|
Crezee T, Tesselaar MH, Nagarajah J, Corver WE, Morreau J, Pritchard C, Kimura S, Kuiper JG, van Engen-van Grunsven I, Smit JWA, Netea-Maier RT, Plantinga TS. Digoxin treatment reactivates in vivo radioactive iodide uptake and correlates with favorable clinical outcome in non-medullary thyroid cancer. Cell Oncol (Dordr) 2021; 44:611-625. [PMID: 33534128 PMCID: PMC8213564 DOI: 10.1007/s13402-021-00588-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Non-medullary thyroid cancer (NMTC) treatment is based on the ability of thyroid follicular cells to accumulate radioactive iodide (RAI). However, in a subset of NMTC patients tumor dedifferentiation occurs, leading to RAI resistance. Digoxin has been demonstrated to restore iodide uptake capacity in vitro in poorly differentiated and anaplastic NMTC cells, termed redifferentiation. The aim of the present study was to investigate the in vivo effects of digoxin in TPO-Cre/LSL-BrafV600E mice and digoxin-treated NMTC patients. METHODS Mice with thyroid cancer were subjected to 3D ultrasound for monitoring tumor growth and 124I PET/CT for measurement of intratumoral iodide uptake. Post-mortem analyses on tumor tissues comprised gene expression profiling and measurement of intratumoral autophagy activity. Through PALGA (Dutch Pathology Registry), archived tumor material was obtained from 11 non-anaplastic NMTC patients who were using digoxin. Clinical characteristics and tumor material of these patients were compared to 11 matched control NMTC patients never treated with digoxin. RESULTS We found that in mice, tumor growth was inhibited and 124I accumulation was sustainably increased after short-course digoxin treatment. Post-mortem analyses revealed that digoxin treatment increased autophagy activity and enhanced expression of thyroid-specific genes in mouse tumors compared to vehicle-treated mice. Digoxin-treated NMTC patients exhibited significantly higher autophagy activity and a higher differentiation status as compared to matched control NMTC patients, and were associated with favourable clinical outcome. CONCLUSIONS These in vivo data support the hypothesis that digoxin may represent a repositioned adjunctive treatment modality that suppresses tumor growth and improves RAI sensitivity in patients with RAI-refractory NMTC.
Collapse
Affiliation(s)
- Thomas Crezee
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands.
| | - Marika H Tesselaar
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| | - James Nagarajah
- Department of Radiology & Nuclear Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Willem E Corver
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catrin Pritchard
- Department of Pathology, University of Leicester, Leicester, LEI7RH, UK
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Ilse van Engen-van Grunsven
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| | - Jan W A Smit
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Theo S Plantinga
- Department of Pathology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 10, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
52
|
Soleimanpour E, Bergado Acosta JR, Landgraf P, Mayer D, Dankert E, Dieterich DC, Fendt M. Regulation of CREB Phosphorylation in Nucleus Accumbens after Relief Conditioning. Cells 2021; 10:238. [PMID: 33530478 PMCID: PMC7912172 DOI: 10.3390/cells10020238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 11/16/2022] Open
Abstract
Relief learning is the association of environmental cues with the cessation of aversive events. While there is increasing knowledge about the neural circuitry mediating relief learning, the respective molecular pathways are not known. Therefore, the aim of the present study was to examine different putative molecular pathways underlying relief learning. To this purpose, male rats were subjected either to relief conditioning or to a pseudo conditioning procedure. Forty-five minutes or 6 h after conditioning, samples of five different brain regions, namely the prefrontal cortex, nucleus accumbens (NAC), dorsal striatum, dorsal hippocampus, and amygdala, were collected. Using quantitative Western blots, the expression level of CREB, pCREB, ERK1/2, pERK1/2, CaMKIIα, MAP2K, PKA, pPKA, Akt, pAkt, DARPP-32, pDARPP-32, 14-3-3, and neuroligin2 were studied. Our analyses revealed that relief conditioned rats had higher CREB phosphorylation in NAC 6 h after conditioning than pseudo conditioned rats. The data further revealed that this CREB phosphorylation was mainly induced by dopamine D1 receptor-mediated activation of PKA, however, other kinases, downstream of the NMDA receptor, may also contribute. Taken together, the present study suggests that CREB phosphorylation, induced by a combination of different molecular pathways downstream of dopamine D1 and NMDA receptors, is essential for the acquisition and consolidation of relief learning.
Collapse
Affiliation(s)
- Elaheh Soleimanpour
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
| | - Jorge R. Bergado Acosta
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
- Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Peter Landgraf
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
| | - Dana Mayer
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
| | - Evelyn Dankert
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
| | - Daniela C. Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
- Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (J.R.B.A.); (P.L.); (D.M.); (E.D.); (D.C.D.); (M.F.)
- Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, 39106 Magdeburg, Germany
| |
Collapse
|
53
|
Ren B, Schmid M, Scheiner M, Mollenkopf HJ, Lucius R, Heitlinger E, Gupta N. Toxoplasma and Eimeria co-opt the host cFos expression for intracellular development in mammalian cells. Comput Struct Biotechnol J 2021; 19:719-731. [PMID: 33510872 PMCID: PMC7817532 DOI: 10.1016/j.csbj.2020.12.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/05/2022] Open
Abstract
Gene expression profiles differ significantly between Toxoplasma and Eimeria-infected host cells. Several distinct and shared host-signaling cascades are regulated by coccidian parasites. cFos is one of the few host transcripts mutually regulated during infection by both pathogens. Host cFos is required for optimal in vitro development of E. falciformis and T. gondii. Transcriptomics of parasitized wild-type and cFos-/- host cells reveals a perturbation of cFos network.
Successful asexual reproduction of intracellular pathogens depends on their potential to exploit host resources and subvert antimicrobial defense. In this work, we deployed two prevalent apicomplexan parasites of mammalian cells, namely Toxoplasma gondii and Eimeria falciformis, to identify potential host determinants of infection. Expression analyses of the young adult mouse colonic (YAMC) epithelial cells upon infection by either parasite showed regulation of several distinct transcripts, indicating that these two pathogens program their intracellular niches in a tailored manner. Conversely, parasitized mouse embryonic fibroblasts (MEFs) displayed a divergent transcriptome compared to corresponding YAMC epithelial cells, suggesting that individual host cells mount a fairly discrete response when encountering a particular pathogen. Among several host transcripts similarly altered by T. gondii and E. falciformis, we identified cFos, a master transcription factor, that was consistently induced throughout the infection. Indeed, asexual growth of both parasites was strongly impaired in MEF host cells lacking cFos expression. Last but not the least, our differential transcriptomics of the infected MEFs (parental and cFos-/- mutant) and YAMC epithelial cells disclosed a cFos-centered network, underlying signal cascades, as well as a repertoire of nucleotides- and ion-binding proteins, which presumably act in consort to acclimatize the mammalian cell and thereby facilitate the parasite development.
Collapse
Affiliation(s)
- Bingjian Ren
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Manuela Schmid
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Mattea Scheiner
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Hans-Joachim Mollenkopf
- Microarray and Genomics Core Facility, Max-Planck Institute for Infection Biology, Berlin, Germany
| | - Richard Lucius
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Emanuel Heitlinger
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Research Group Ecology and Evolution of Parasite Host Interactions, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Nishith Gupta
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Department of Biological Sciences, Birla Institute of Technology and Science Pilani (BITS-P), Hyderabad, India
| |
Collapse
|
54
|
Pfeil EM, Brands J, Merten N, Vögtle T, Vescovo M, Rick U, Albrecht IM, Heycke N, Kawakami K, Ono Y, Ngako Kadji FM, Hiratsuka S, Aoki J, Häberlein F, Matthey M, Garg J, Hennen S, Jobin ML, Seier K, Calebiro D, Pfeifer A, Heinemann A, Wenzel D, König GM, Nieswandt B, Fleischmann BK, Inoue A, Simon K, Kostenis E. Heterotrimeric G Protein Subunit Gαq Is a Master Switch for Gβγ-Mediated Calcium Mobilization by Gi-Coupled GPCRs. Mol Cell 2020; 80:940-954.e6. [PMID: 33202251 DOI: 10.1016/j.molcel.2020.10.027] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Mechanisms that control mobilization of cytosolic calcium [Ca2+]i are key for regulation of numerous eukaryotic cell functions. One such paradigmatic mechanism involves activation of phospholipase Cβ (PLCβ) enzymes by G protein βγ subunits from activated Gαi-Gβγ heterotrimers. Here, we report identification of a master switch to enable this control for PLCβ enzymes in living cells. We find that the Gαi-Gβγ-PLCβ-Ca2+ signaling module is entirely dependent on the presence of active Gαq. If Gαq is pharmacologically inhibited or genetically ablated, Gβγ can bind to PLCβ but does not elicit Ca2+ signals. Removal of an auto-inhibitory linker that occludes the active site of the enzyme is required and sufficient to empower "stand-alone control" of PLCβ by Gβγ. This dependence of Gi-Gβγ-Ca2+ on Gαq places an entire signaling branch of G-protein-coupled receptors (GPCRs) under hierarchical control of Gq and changes our understanding of how Gi-GPCRs trigger [Ca2+]i via PLCβ enzymes.
Collapse
Affiliation(s)
- Eva Marie Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Julian Brands
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Timo Vögtle
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Maddalena Vescovo
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Ulrike Rick
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Ina-Maria Albrecht
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Yuki Ono
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | | | - Suzune Hiratsuka
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Felix Häberlein
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jaspal Garg
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Stephanie Hennen
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Marie-Lise Jobin
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, 97078 Würzburg, Germany
| | - Kerstin Seier
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, 97078 Würzburg, Germany
| | - Davide Calebiro
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, 97078 Würzburg, Germany; Institute of Metabolism and Systems Research and Centre of Membrane Proteins and Receptors, University of Birmingham, B15 2TT Birmingham, UK
| | - Alexander Pfeifer
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Akos Heinemann
- Division of Pharmacology, Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, 8010 Graz, Austria
| | - Daniela Wenzel
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany.
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany.
| |
Collapse
|
55
|
Aponte-Santiago NA, Littleton JT. Synaptic Properties and Plasticity Mechanisms of Invertebrate Tonic and Phasic Neurons. Front Physiol 2020; 11:611982. [PMID: 33391026 PMCID: PMC7772194 DOI: 10.3389/fphys.2020.611982] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Defining neuronal cell types and their associated biophysical and synaptic diversity has become an important goal in neuroscience as a mechanism to create comprehensive brain cell atlases in the post-genomic age. Beyond broad classification such as neurotransmitter expression, interneuron vs. pyramidal, sensory or motor, the field is still in the early stages of understanding closely related cell types. In both vertebrate and invertebrate nervous systems, one well-described distinction related to firing characteristics and synaptic release properties are tonic and phasic neuronal subtypes. In vertebrates, these classes were defined based on sustained firing responses during stimulation (tonic) vs. transient responses that rapidly adapt (phasic). In crustaceans, the distinction expanded to include synaptic release properties, with tonic motoneurons displaying sustained firing and weaker synapses that undergo short-term facilitation to maintain muscle contraction and posture. In contrast, phasic motoneurons with stronger synapses showed rapid depression and were recruited for short bursts during fast locomotion. Tonic and phasic motoneurons with similarities to those in crustaceans have been characterized in Drosophila, allowing the genetic toolkit associated with this model to be used for dissecting the unique properties and plasticity mechanisms for these neuronal subtypes. This review outlines general properties of invertebrate tonic and phasic motoneurons and highlights recent advances that characterize distinct synaptic and plasticity pathways associated with two closely related glutamatergic neuronal cell types that drive invertebrate locomotion.
Collapse
Affiliation(s)
- Nicole A. Aponte-Santiago
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - J. Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
56
|
Jedrychowska J, Gasanov EV, Korzh V. Kcnb1 plays a role in development of the inner ear. Dev Biol 2020; 471:65-75. [PMID: 33316259 DOI: 10.1016/j.ydbio.2020.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
The function of the inner ear depends on the maintenance of high concentrations of K+ ions. The slow-inactivating delayed rectifier Kv2.1/KCNB1 channel works in the inner ear in mammals. The kcnb1 gene is expressed in the otic vesicle of developing zebrafish, suggesting its role in development of the inner ear. In the present study, we found that a Kcnb1 loss-of-function mutation affected development of the inner ear at multiple levels, including otic vesicle expansion, otolith formation, and the proliferation and differentiation of mechanosensory cells. This resulted in defects of kinocilia and stereocilia and abnormal function of the inner ear detected by behavioral assays. The quantitative transcriptional analysis of 75 genes demonstrated that the kcnb1 mutation affected the transcription of genes that are involved in K+ metabolism, cell proliferation, cilia development, and intracellular protein trafficking. These results demonstrate a role for Kv2.1/Kcnb1 channels in development of the inner ear in zebrafish.
Collapse
Affiliation(s)
- Justyna Jedrychowska
- International Institute of Molecular and Cell Biology in Warsaw, Poland; Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Eugene V Gasanov
- International Institute of Molecular and Cell Biology in Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Poland.
| |
Collapse
|
57
|
Adiga D, Radhakrishnan R, Chakrabarty S, Kumar P, Kabekkodu SP. The Role of Calcium Signaling in Regulation of Epithelial-Mesenchymal Transition. Cells Tissues Organs 2020; 211:134-156. [PMID: 33316804 DOI: 10.1159/000512277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/13/2020] [Indexed: 11/19/2022] Open
Abstract
Despite substantial advances in the field of cancer therapeutics, metastasis is a significant challenge for a favorable clinical outcome. Epithelial to mesenchymal transition (EMT) is a process of acquiring increased motility, invasiveness, and therapeutic resistance by cancer cells for their sustained growth and survival. A plethora of intrinsic mechanisms and extrinsic microenvironmental factors drive the process of cancer metastasis. Calcium (Ca2+) signaling plays a critical role in dictating the adaptive metastatic cell behavior comprising of cell migration, invasion, angiogenesis, and intravasation. By modulating EMT, Ca2+ signaling can regulate the complexity and dynamics of events leading to metastasis. This review summarizes the role of Ca2+ signal remodeling in the regulation of EMT and metastasis in cancer.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India
| | - Prashant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India,
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India,
| |
Collapse
|
58
|
Chen H, Vandorpe DH, Xie X, Alper SL, Zeidel ML, Yu W. Disruption of Cav1.2-mediated signaling is a pathway for ketamine-induced pathology. Nat Commun 2020; 11:4328. [PMID: 32859919 PMCID: PMC7455701 DOI: 10.1038/s41467-020-18167-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/04/2020] [Indexed: 01/03/2023] Open
Abstract
The general anesthetic ketamine has been repurposed by physicians as an anti-depressant and by the public as a recreational drug. However, ketamine use can cause extensive pathological changes, including ketamine cystitis. The mechanisms of ketamine's anti-depressant and adverse effects remain poorly understood. Here we present evidence that ketamine is an effective L-type Ca2+ channel (Cav1.2) antagonist that directly inhibits calcium influx and smooth muscle contractility, leading to voiding dysfunction. Ketamine prevents Cav1.2-mediated induction of immediate early genes and transcription factors, and inactivation of Cav1.2 in smooth muscle mimics the ketamine cystitis phenotype. Our results demonstrate that ketamine inhibition of Cav1.2 signaling is an important pathway mediating ketamine cystitis. In contrast, Cav1.2 agonist Bay k8644 abrogates ketamine-induced smooth muscle dysfunction. Indeed, Cav1.2 activation by Bay k8644 decreases voiding frequency while increasing void volume, indicating Cav1.2 agonists might be effective drugs for treatment of bladder dysfunction.
Collapse
Affiliation(s)
- Huan Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David H Vandorpe
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xiang Xie
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Seth L Alper
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark L Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Weiqun Yu
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
59
|
Tripathi MK, Kartawy M, Amal H. The role of nitric oxide in brain disorders: Autism spectrum disorder and other psychiatric, neurological, and neurodegenerative disorders. Redox Biol 2020; 34:101567. [PMID: 32464501 PMCID: PMC7256645 DOI: 10.1016/j.redox.2020.101567] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) is a multifunctional signalling molecule and a neurotransmitter that plays an important role in physiological and pathophysiological processes. In physiological conditions, NO regulates cell survival, differentiation and proliferation of neurons. It also regulates synaptic activity, plasticity and vesicle trafficking. NO affects cellular signalling through protein S-nitrosylation, the NO-mediated posttranslational modification of cysteine thiols (SNO). SNO can affect protein activity, protein-protein interaction and protein localization. Numerous studies have shown that excessive NO and SNO can lead to nitrosative stress in the nervous system, contributing to neuropathology. In this review, we summarize the role of NO and SNO in the progression of neurodevelopmental, psychiatric and neurodegenerative disorders, with special attention to autism spectrum disorder (ASD). We provide mechanistic insights into the contribution of NO in diverse brain disorders. Finally, we suggest that pharmacological agents that can inhibit or augment the production of NO as well as new approaches to modulate the formation of SNO-proteins can serve as a promising approach for the treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
60
|
Deletion of Voltage-Gated Calcium Channels in Astrocytes during Demyelination Reduces Brain Inflammation and Promotes Myelin Regeneration in Mice. J Neurosci 2020; 40:3332-3347. [PMID: 32169969 DOI: 10.1523/jneurosci.1644-19.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/28/2022] Open
Abstract
To determine whether Cav1.2 voltage-gated Ca2+ channels contribute to astrocyte activation, we generated an inducible conditional knock-out mouse in which the Cav1.2 α subunit was deleted in GFAP-positive astrocytes. This astrocytic Cav1.2 knock-out mouse was tested in the cuprizone model of myelin injury and repair which causes astrocyte and microglia activation in the absence of a lymphocytic response. Deletion of Cav1.2 channels in GFAP-positive astrocytes during cuprizone-induced demyelination leads to a significant reduction in the degree of astrocyte and microglia activation and proliferation in mice of either sex. Concomitantly, the production of proinflammatory factors such as TNFα, IL1β and TGFβ1 was significantly decreased in the corpus callosum and cortex of Cav1.2 knock-out mice through demyelination. Furthermore, this mild inflammatory environment promotes oligodendrocyte progenitor cells maturation and myelin regeneration across the remyelination phase of the cuprizone model. Similar results were found in animals treated with nimodipine, a Cav1.2 Ca2+ channel inhibitor with high affinity to the CNS. Mice of either sex injected with nimodipine during the demyelination stage of the cuprizone treatment displayed a reduced number of reactive astrocytes and showed a faster and more efficient brain remyelination. Together, these results indicate that Cav1.2 Ca2+ channels play a crucial role in the induction and proliferation of reactive astrocytes during demyelination; and that attenuation of astrocytic voltage-gated Ca2+ influx may be an effective therapy to reduce brain inflammation and promote myelin recovery in demyelinating diseases.SIGNIFICANCE STATEMENT Reducing voltage-gated Ca2+ influx in astrocytes during brain demyelination significantly attenuates brain inflammation and astrocyte reactivity. Furthermore, these changes promote myelin restoration and oligodendrocyte maturation throughout remyelination.
Collapse
|
61
|
Havis E, Duprez D. EGR1 Transcription Factor is a Multifaceted Regulator of Matrix Production in Tendons and Other Connective Tissues. Int J Mol Sci 2020; 21:ijms21051664. [PMID: 32121305 PMCID: PMC7084410 DOI: 10.3390/ijms21051664] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022] Open
Abstract
Although the transcription factor EGR1 is known as NGF1-A, TIS8, Krox24, zif/268, and ZENK, it still has many fewer names than biological functions. A broad range of signals induce Egr1 gene expression via numerous regulatory elements identified in the Egr1 promoter. EGR1 is also the target of multiple post-translational modifications, which modulate EGR1 transcriptional activity. Despite the myriad regulators of Egr1 transcription and translation, and the numerous biological functions identified for EGR1, the literature reveals a recurring theme of EGR1 transcriptional activity in connective tissues, regulating genes related to the extracellular matrix. Egr1 is expressed in different connective tissues, such as tendon (a dense connective tissue), cartilage and bone (supportive connective tissues), and adipose tissue (a loose connective tissue). Egr1 is involved in the development, homeostasis, and healing processes of these tissues, mainly via the regulation of extracellular matrix. In addition, Egr1 is often involved in the abnormal production of extracellular matrix in fibrotic conditions, and Egr1 deletion is seen as a target for therapeutic strategies to fight fibrotic conditions. This generic EGR1 function in matrix regulation has little-explored implications but is potentially important for tendon repair.
Collapse
|
62
|
Ohata J, Krishnamoorthy L, Gonzalez MA, Xiao T, Iovan DA, Toste FD, Miller EW, Chang CJ. An Activity-Based Methionine Bioconjugation Approach To Developing Proximity-Activated Imaging Reporters. ACS CENTRAL SCIENCE 2020; 6:32-40. [PMID: 31989024 PMCID: PMC6978837 DOI: 10.1021/acscentsci.9b01038] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Indexed: 05/04/2023]
Abstract
Chemical probes that report on protein activity, rather than protein abundance, with spatial and temporal resolution can enable studies of their native function in biological contexts as well as provide opportunities for developing new types of biochemical reporters. Here we present a sensing platform, termed proximity-activated imaging reporter (PAIR), which combines activity-based methionine bioconjugation and antibody labeling with proximity-dependent oligonucleotide-based amplification to monitor dynamic changes of a given analyte in cells and animals through context-dependent methionine labeling of specific protein targets. We establish this PAIR method to develop sensors for imaging reactive oxygen species (ROS) and calcium ions through oxaziridine-directed labeling of reactive methionine residues on β-actin and calmodulin (CaM), respectively, where the extent of methionine bioconjugation on these protein targets can serve as an indicator of oxidative stress or calcium status. In particular, application of PAIR to activity-based CaM detection provides a method for imaging integrated calcium activity in both in vitro cell and in vivo zebrafish models. By relying on native protein biochemistry, PAIR enables redox and metal imaging without introduction of external small molecules or genetically encoded indicators that can potentially buffer the natural/existing pools. This approach can be potentially generalized to target a broader range of analytes by pairing appropriate activity-based protein probes with protein detection reagents in a proximity-driven manner, providing a starting point not only for designing new sensors but also for monitoring endogenous activity of specific protein targets in biological specimens with spatial and temporal fidelity.
Collapse
Affiliation(s)
- Jun Ohata
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Lakshmi Krishnamoorthy
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Monica A. Gonzalez
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Tong Xiao
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Diana A. Iovan
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - F. Dean Toste
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Evan W. Miller
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| | - Christopher J. Chang
- Department
of Chemistry, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
| |
Collapse
|
63
|
Avazzadeh S, McDonagh K, Reilly J, Wang Y, Boomkamp SD, McInerney V, Krawczyk J, Fitzgerald J, Feerick N, O'Sullivan M, Jalali A, Forman EB, Lynch SA, Ennis S, Cosemans N, Peeters H, Dockery P, O'Brien T, Quinlan LR, Gallagher L, Shen S. Increased Ca 2+ signaling in NRXN1α +/- neurons derived from ASD induced pluripotent stem cells. Mol Autism 2019; 10:52. [PMID: 31893021 PMCID: PMC6937972 DOI: 10.1186/s13229-019-0303-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/05/2019] [Indexed: 12/28/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a high co-morbidity of epilepsy and associated with hundreds of rare risk factors. NRXN1 deletion is among the commonest rare genetic factors shared by ASD, schizophrenia, intellectual disability, epilepsy, and developmental delay. However, how NRXN1 deletions lead to different clinical symptoms is unknown. Patient-derived cells are essential to investigate the functional consequences of NRXN1 lesions to human neurons in different diseases. Methods Skin biopsies were donated by five healthy donors and three ASD patients carrying NRXN1α+/− deletions. Seven control and six NRXN1α+/− iPSC lines were derived and differentiated into day 100 cortical excitatory neurons using dual SMAD inhibition. Calcium (Ca2+) imaging was performed using Fluo4-AM, and the properties of Ca2+ transients were compared between two groups of neurons. Transcriptome analysis was carried out to undercover molecular pathways associated with NRXN1α+/− neurons. Results NRXN1α+/− neurons were found to display altered calcium dynamics, with significantly increased frequency, duration, and amplitude of Ca2+ transients. Whole genome RNA sequencing also revealed altered ion transport and transporter activity, with upregulated voltage-gated calcium channels as one of the most significant pathways in NRXN1α+/− neurons identified by STRING and GSEA analyses. Conclusions This is the first report to show that human NRXN1α+/− neurons derived from ASD patients’ iPSCs present novel phenotypes of upregulated VGCCs and increased Ca2+ transients, which may facilitate the development of drug screening assays for the treatment of ASD.
Collapse
Affiliation(s)
- Sahar Avazzadeh
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Katya McDonagh
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Jamie Reilly
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Yanqin Wang
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland.,2Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Stephanie D Boomkamp
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Veronica McInerney
- 3HRB Clinical Research Facility, National University of Ireland (NUI), Galway, Ireland
| | - Janusz Krawczyk
- 4Department of Haematology, Galway University Hospital, Galway, Ireland
| | | | - Niamh Feerick
- 5School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Amirhossein Jalali
- 6School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eva B Forman
- 7Children's University Hospital, Temple Street, Dublin, Ireland
| | - Sally A Lynch
- Department of Clinical Genetics, OLCHC, Dublin 12, Ireland.,9Children's University Hospital, Temple St, Dublin, Ireland.,10Academic Center on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Sean Ennis
- 11UCD Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Nele Cosemans
- 12Centre for Human Genetics, University Hospital Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Peeters
- 10Academic Center on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Peter Dockery
- 13Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | - Timothy O'Brien
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Leo R Quinlan
- 14Physiology and Human Movement Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | | | - Sanbing Shen
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| |
Collapse
|
64
|
Moretti R, Peinkhofer C. B Vitamins and Fatty Acids: What Do They Share with Small Vessel Disease-Related Dementia? Int J Mol Sci 2019; 20:5797. [PMID: 31752183 PMCID: PMC6888477 DOI: 10.3390/ijms20225797] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/21/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Many studies have been written on vitamin supplementation, fatty acid, and dementia, but results are still under debate, and no definite conclusion has yet been drawn. Nevertheless, a significant amount of lab evidence confirms that vitamins of the B group are tightly related to gene control for endothelium protection, act as antioxidants, play a co-enzymatic role in the most critical biochemical reactions inside the brain, and cooperate with many other elements, such as choline, for the synthesis of polyunsaturated phosphatidylcholine, through S-adenosyl-methionine (SAM) methyl donation. B-vitamins have anti-inflammatory properties and act in protective roles against neurodegenerative mechanisms, for example, through modulation of the glutamate currents and a reduction of the calcium currents. In addition, they also have extraordinary antioxidant properties. However, laboratory data are far from clinical practice. Many studies have tried to apply these results in everyday clinical activity, but results have been discouraging and far from a possible resolution of the associated mysteries, like those represented by Alzheimer's disease (AD) or small vessel disease dementia. Above all, two significant problems emerge from the research: No consensus exists on general diagnostic criteria-MCI or AD? Which diagnostic criteria should be applied for small vessel disease-related dementia? In addition, no general schema exists for determining a possible correct time of implementation to have effective results. Here we present an up-to-date review of the literature on such topics, shedding some light on the possible interaction of vitamins and phosphatidylcholine, and their role in brain metabolism and catabolism. Further studies should take into account all of these questions, with well-designed and world-homogeneous trials.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
65
|
Roles for the Endoplasmic Reticulum in Regulation of Neuronal Calcium Homeostasis. Cells 2019; 8:cells8101232. [PMID: 31658749 PMCID: PMC6829861 DOI: 10.3390/cells8101232] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
By influencing Ca2+ homeostasis in spatially and architecturally distinct neuronal compartments, the endoplasmic reticulum (ER) illustrates the notion that form and function are intimately related. The contribution of ER to neuronal Ca2+ homeostasis is attributed to the organelle being the largest reservoir of intracellular Ca2+ and having a high density of Ca2+ channels and transporters. As such, ER Ca2+ has incontrovertible roles in the regulation of axodendritic growth and morphology, synaptic vesicle release, and neural activity dependent gene expression, synaptic plasticity, and mitochondrial bioenergetics. Not surprisingly, many neurological diseases arise from ER Ca2+ dyshomeostasis, either directly due to alterations in ER resident proteins, or indirectly via processes that are coupled to the regulators of ER Ca2+ dynamics. In this review, we describe the mechanisms involved in the establishment of ER Ca2+ homeostasis in neurons. We elaborate upon how changes in the spatiotemporal dynamics of Ca2+ exchange between the ER and other organelles sculpt neuronal function and provide examples that demonstrate the involvement of ER Ca2+ dyshomeostasis in a range of neurological and neurodegenerative diseases.
Collapse
|
66
|
Brigidi GS, Hayes MGB, Delos Santos NP, Hartzell AL, Texari L, Lin PA, Bartlett A, Ecker JR, Benner C, Heinz S, Bloodgood BL. Genomic Decoding of Neuronal Depolarization by Stimulus-Specific NPAS4 Heterodimers. Cell 2019; 179:373-391.e27. [PMID: 31585079 PMCID: PMC6800120 DOI: 10.1016/j.cell.2019.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/22/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
Cells regulate gene expression in response to salient external stimuli. In neurons, depolarization leads to the expression of inducible transcription factors (ITFs) that direct subsequent gene regulation. Depolarization encodes both a neuron's action potential (AP) output and synaptic inputs, via excitatory postsynaptic potentials (EPSPs). However, it is unclear if distinct types of electrical activity can be transformed by an ITF into distinct modes of genomic regulation. Here, we show that APs and EPSPs in mouse hippocampal neurons trigger two spatially segregated and molecularly distinct induction mechanisms that lead to the expression of the ITF NPAS4. These two pathways culminate in the formation of stimulus-specific NPAS4 heterodimers that exhibit distinct DNA binding patterns. Thus, NPAS4 differentially communicates increases in a neuron's spiking output and synaptic inputs to the nucleus, enabling gene regulation to be tailored to the type of depolarizing activity along the somato-dendritic axis of a neuron.
Collapse
Affiliation(s)
- G Stefano Brigidi
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Michael G B Hayes
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Nathaniel P Delos Santos
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA; Department of Biomedical Informatics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Andrea L Hartzell
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA; Neuroscience Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Lorane Texari
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Pei-Ann Lin
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA; Neuroscience Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, La Jolla, CA 92093, USA
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Sven Heinz
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA
| | - Brenda L Bloodgood
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634, USA.
| |
Collapse
|
67
|
Calcineurin controls gene transcription following stimulation of a Gαq-coupled designer receptor. Exp Cell Res 2019; 383:111553. [DOI: 10.1016/j.yexcr.2019.111553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/07/2019] [Accepted: 08/10/2019] [Indexed: 01/07/2023]
|
68
|
Weathington N, O’Brien ME, Radder J, Whisenant TC, Bleecker ER, Busse WW, Erzurum SC, Gaston B, Hastie AT, Jarjour NN, Meyers DA, Milosevic J, Moore WC, Tedrow JR, Trudeau JB, Wong HP, Wu W, Kaminski N, Wenzel SE, Modena BD. BAL Cell Gene Expression in Severe Asthma Reveals Mechanisms of Severe Disease and Influences of Medications. Am J Respir Crit Care Med 2019; 200:837-856. [PMID: 31161938 PMCID: PMC6812436 DOI: 10.1164/rccm.201811-2221oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/03/2019] [Indexed: 01/16/2023] Open
Abstract
Rationale: Gene expression of BAL cells, which samples the cellular milieu within the lower respiratory tract, has not been well studied in severe asthma.Objectives: To identify new biomolecular mechanisms underlying severe asthma by an unbiased, detailed interrogation of global gene expression.Methods: BAL cell expression was profiled in 154 asthma and control subjects. Of these participants, 100 had accompanying airway epithelial cell gene expression. BAL cell expression profiles were related to participant (age, sex, race, and medication) and sample traits (cell proportions), and then severity-related gene expression determined by correlating transcripts and coexpression networks to lung function, emergency department visits or hospitalizations in the last year, medication use, and quality-of-life scores.Measurements and Main Results: Age, sex, race, cell proportions, and medications strongly influenced BAL cell gene expression, but leading severity-related genes could be determined by carefully identifying and accounting for these influences. A BAL cell expression network enriched for cAMP signaling components most differentiated subjects with severe asthma from other subjects. Subsequently, an in vitro cellular model showed this phenomenon was likely caused by a robust upregulation in cAMP-related expression in nonsevere and β-agonist-naive subjects given a β-agonist before cell collection. Interestingly, ELISAs performed on BAL lysates showed protein levels may partly disagree with expression changes.Conclusions: Gene expression in BAL cells is influenced by factors seldomly considered. Notably, β-agonist exposure likely had a strong and immediate impact on cellular gene expression, which may not translate to important disease mechanisms or necessarily match protein levels. Leading severity-related genes were discovered in an unbiased, system-wide analysis, revealing new targets that map to asthma susceptibility loci.
Collapse
Affiliation(s)
- Nathaniel Weathington
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael E. O’Brien
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Josiah Radder
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas C. Whisenant
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Eugene R. Bleecker
- Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, Arizona
| | - William W. Busse
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin, Madison, Wisconsin
| | - Serpil C. Erzurum
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Benjamin Gaston
- Division of Pediatric Pulmonary, Allergy and Immunology, Case Western Reserve University and Rainbow Babies Children’s Hospital, Cleveland, Ohio
| | - Annette T. Hastie
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Nizar N. Jarjour
- Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin, Madison, Wisconsin
| | - Deborah A. Meyers
- Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, Arizona
| | - Jadranka Milosevic
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wendy C. Moore
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John R. Tedrow
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John B. Trudeau
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hesper P. Wong
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wei Wu
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Sally E. Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Brian D. Modena
- Division of Allergy, National Jewish Hospital, Denver, Colorado
| |
Collapse
|
69
|
Hao Y, Wang L, Chen H, Hill WG, Robson SC, Zeidel ML, Yu W. Targetable purinergic receptors P2Y12 and A2b antagonistically regulate bladder function. JCI Insight 2019; 4:122112. [PMID: 31434806 DOI: 10.1172/jci.insight.122112] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/26/2019] [Indexed: 12/21/2022] Open
Abstract
Abnormalities in purine availability or purinergic receptor density are commonly seen in patients with lower urinary tract symptoms (LUTS), but the underlying mechanisms relating altered receptor function to LUTS are unknown. Here we provide extensive evidence for the reciprocal interplay of multiple receptors responding to ATP, ADP (adenosine diphosphate), and adenosine, agonists that regulate bladder function significantly. ADP stimulated P2Y12 receptors, causing bladder smooth muscle (BSM) contraction, whereas adenosine signaling through potentially newly defined A2b receptors, actively inhibited BSM purinergic contractility. The modulation of adenylyl cyclase-cAMP signaling via A2b and P2Y12 interaction actively regulated bladder contractility by modulating intracellular calcium levels. KO mice lacking the receptors display diametrically opposed bladder phenotypes, with P2Y12-KO mice exhibiting an underactive bladder (UAB) phenotype with increased bladder capacity and reduced voiding frequency, whereas A2b-KO mice have an overactive bladder (OAB), with decreased capacity and increased voiding frequency. The opposing phenotypes in P2Y12-KO and A2b-KO mice not only resulted from dysregulated BSM contractility, but also from abnormal BSM cell growth. Finally, we demonstrate that i.p. administration of drugs targeting P2Y12 or A2b receptor rescues these abnormal phenotypes in both KO mice. These findings strongly indicate that P2Y12 and A2b receptors are attractive therapeutic targets for human patients with LUTS.
Collapse
Affiliation(s)
- Yuan Hao
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Lu Wang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Chongqing University, Chongqing, China
| | - Huan Chen
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Warren G Hill
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark L Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Weiqun Yu
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
70
|
Tyebji S, Seizova S, Garnham AL, Hannan AJ, Tonkin CJ. Impaired social behaviour and molecular mediators of associated neural circuits during chronic Toxoplasma gondii infection in female mice. Brain Behav Immun 2019; 80:88-108. [PMID: 30807837 DOI: 10.1016/j.bbi.2019.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/22/2019] [Indexed: 12/24/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a neurotropic parasite that is associated with various neuropsychiatric disorders. Rodents infected with T. gondii display a plethora of behavioural alterations, and Toxoplasma infection in humans has been strongly associated with disorders such as schizophrenia, in which impaired social behaviour is an important feature. Elucidating changes at the cellular level relevant to neuropsychiatric conditions can lead to effective therapies. Here, we compare changes in behaviour during an acute and chronic T. gondii infection in female mice. Further, we notice that during chronic phase of infection, mice display impaired sociability when exposed to a novel conspecific. Also, we show that T. gondii infected mice display impaired short-term social recognition memory. However, object recognition memory remains intact. Using c-Fos as a marker of neuronal activity, we show that infection leads to an impairment in neuronal activation in the medial prefrontal cortex, hippocampus as well as the amygdala when mice are exposed to a social environment and a change in functional connectivity between these regions. We found changes in synaptic proteins that play a role in the process of neuronal activation such as synaptophysin, PSD-95 and changes in downstream substrates of cell activity such as cyclic AMP, phospho-CREB and BDNF. Our results point towards an imbalance in neuronal activity that can lead to a wider range of neuropsychiatric problems upon T. gondii infection.
Collapse
Affiliation(s)
- Shiraz Tyebji
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia.
| | - Simona Seizova
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville 3052, Victoria, Australia.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| |
Collapse
|
71
|
Rapid and active stabilization of visual cortical firing rates across light-dark transitions. Proc Natl Acad Sci U S A 2019; 116:18068-18077. [PMID: 31366632 DOI: 10.1073/pnas.1906595116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The dynamics of neuronal firing during natural vision are poorly understood. Surprisingly, mean firing rates of neurons in primary visual cortex (V1) of freely behaving rodents are similar during prolonged periods of light and darkness, but it is unknown whether this reflects a slow adaptation to changes in natural visual input or insensitivity to rapid changes in visual drive. Here, we use chronic electrophysiology in freely behaving rats to follow individual V1 neurons across many dark-light (D-L) and light-dark (L-D) transitions. We show that, even on rapid timescales (1 s to 10 min), neuronal activity was only weakly modulated by transitions that coincided with the expected 12-/12-h L-D cycle. In contrast, a larger subset of V1 neurons consistently responded to unexpected L-D and D-L transitions, and disruption of the regular L-D cycle with 60 h of complete darkness induced a robust increase in V1 firing on reintroduction of visual input. Thus, V1 neurons fire at similar rates in the presence or absence of natural stimuli, and significant changes in activity arise only transiently in response to unexpected changes in the visual environment. Furthermore, although mean rates were similar in light and darkness, pairwise correlations were significantly stronger during natural vision, suggesting that information about natural scenes in V1 may be more strongly reflected in correlations than individual firing rates. Together, our findings show that V1 firing rates are rapidly and actively stabilized during expected changes in visual input and are remarkably stable at both short and long timescales.
Collapse
|
72
|
Kaldun JC, Sprecher SG. Initiated by CREB: Resolving Gene Regulatory Programs in Learning and Memory. Bioessays 2019; 41:e1900045. [DOI: 10.1002/bies.201900045] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/29/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Jenifer C. Kaldun
- Department of BiologyUniversity of Fribourg1700 Fribourg Switzerland
| | - Simon G. Sprecher
- Department of BiologyUniversity of Fribourg1700 Fribourg Switzerland
| |
Collapse
|
73
|
Matos MR, Visser E, Kramvis I, van der Loo RJ, Gebuis T, Zalm R, Rao-Ruiz P, Mansvelder HD, Smit AB, van den Oever MC. Memory strength gates the involvement of a CREB-dependent cortical fear engram in remote memory. Nat Commun 2019; 10:2315. [PMID: 31127098 PMCID: PMC6534583 DOI: 10.1038/s41467-019-10266-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/29/2019] [Indexed: 12/22/2022] Open
Abstract
Encoding and retrieval of contextual memories is initially mediated by sparsely activated neurons, so-called engram cells, in the hippocampus. Subsequent memory persistence is thought to depend on network-wide changes involving progressive contribution of cortical regions, a process referred to as systems consolidation. Using a viral-based TRAP (targeted recombination in activated populations) approach, we studied whether consolidation of contextual fear memory by neurons in the medial prefrontal cortex (mPFC) is modulated by memory strength and CREB function. We demonstrate that activity of a small subset of mPFC neurons is sufficient and necessary for remote memory expression, but their involvement depends on the strength of conditioning. Furthermore, selective disruption of CREB function in mPFC engram cells after mild conditioning impairs remote memory expression. Together, our data demonstrate that memory consolidation by mPFC engram cells requires CREB-mediated transcription, with the functionality of this network hub being gated by memory strength. Little is known about mechanisms that regulate the involvement of cortical engram cells in remote memory. Here, authors demonstrate that memory consolidation by mPFC engram cells requires CREB-mediated transcription, with the functionality of this network hub being gated by memory strength.
Collapse
Affiliation(s)
- Mariana R Matos
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ioannis Kramvis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Titia Gebuis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Robbert Zalm
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
74
|
Carrano N, Samaddar T, Brunialti E, Franchini L, Marcello E, Ciana P, Mauceri D, Di Luca M, Gardoni F. The Synaptonuclear Messenger RNF10 Acts as an Architect of Neuronal Morphology. Mol Neurobiol 2019; 56:7583-7593. [DOI: 10.1007/s12035-019-1631-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
|
75
|
Pluvinage JV, Haney MS, Smith BAH, Sun J, Iram T, Bonanno L, Li L, Lee DP, Morgens DW, Yang AC, Shuken SR, Gate D, Scott M, Khatri P, Luo J, Bertozzi CR, Bassik MC, Wyss-Coray T. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature 2019; 568:187-192. [PMID: 30944478 PMCID: PMC6574119 DOI: 10.1038/s41586-019-1088-4] [Citation(s) in RCA: 317] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
Abstract
Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris. This function deteriorates during ageing and neurodegenerative disease, concomitant with cognitive decline. However, the mechanisms of impaired microglial homeostatic function and the cognitive effects of restoring this function remain unknown. We combined CRISPR-Cas9 knockout screens with RNA sequencing analysis to discover age-related genetic modifiers of microglial phagocytosis. These screens identified CD22, a canonical B cell receptor, as a negative regulator of phagocytosis that is upregulated on aged microglia. CD22 mediates the anti-phagocytic effect of α2,6-linked sialic acid, and inhibition of CD22 promotes the clearance of myelin debris, amyloid-β oligomers and α-synuclein fibrils in vivo. Long-term central nervous system delivery of an antibody that blocks CD22 function reprograms microglia towards a homeostatic transcriptional state and improves cognitive function in aged mice. These findings elucidate a mechanism of age-related microglial impairment and a strategy to restore homeostasis in the ageing brain.
Collapse
Affiliation(s)
- John V Pluvinage
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Haney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin A H Smith
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Jerry Sun
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Liana Bonanno
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lulin Li
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Davis P Lee
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - David W Morgens
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew C Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Steven R Shuken
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - David Gate
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeleine Scott
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Michael C Bassik
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA.
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
76
|
Sommerlandt FMJ, Brockmann A, Rössler W, Spaethe J. Immediate early genes in social insects: a tool to identify brain regions involved in complex behaviors and molecular processes underlying neuroplasticity. Cell Mol Life Sci 2019; 76:637-651. [PMID: 30349993 PMCID: PMC6514070 DOI: 10.1007/s00018-018-2948-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/25/2018] [Accepted: 10/15/2018] [Indexed: 01/31/2023]
Abstract
Social insects show complex behaviors and master cognitive tasks. The underlying neuronal mechanisms, however, are in most cases only poorly understood due to challenges in monitoring brain activity in freely moving animals. Immediate early genes (IEGs) that get rapidly and transiently expressed following neuronal stimulation provide a powerful tool for detecting behavior-related neuronal activity in vertebrates. In social insects, like honey bees, and in insects in general, this approach is not yet routinely established, even though these genes are highly conserved. First studies revealed a vast potential of using IEGs as neuronal activity markers to analyze the localization, function, and plasticity of neuronal circuits underlying complex social behaviors. We summarize the current knowledge on IEGs in social insects and provide ideas for future research directions.
Collapse
Affiliation(s)
- Frank M J Sommerlandt
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| | - Axel Brockmann
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore, 560065, India
| | - Wolfgang Rössler
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Johannes Spaethe
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| |
Collapse
|
77
|
Cho YK, Park D, Yang A, Chen F, Chuong AS, Klapoetke NC, Boyden ES. Multidimensional screening yields channelrhodopsin variants having improved photocurrent and order-of-magnitude reductions in calcium and proton currents. J Biol Chem 2019; 294:3806-3821. [PMID: 30610117 DOI: 10.1074/jbc.ra118.006996] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Channelrhodopsins (ChRs) are light-gated ion channels in widespread use in neuroscience for mediating the genetically targetable optical control of neurons (optogenetics). ChRs pass multiple kinds of ions, and although nonspecific ChR-mediated conductance is not an issue in many neuroscience studies, conductance of calcium and protons, which can mediate diverse cellular signals, may be undesirable in some instances. Here, we turned our attention to the creation of ChRs that have high cation photocurrent but pass fewer calcium ions and protons. We developed an automated, time-resolved screening method capable of rapidly phenotyping channelrhodopsin-2 (ChR2) variants. We found substitution mutations throughout ChR2 that could boost current while altering ion selectivity and observed that the mutations that reduced calcium or proton conductance have additive effects. By combining four mutations, we obtained a ChR, ChromeQ, with improved photocurrent that possesses order-of-magnitude reductions in calcium and proton conductance and high fidelity in driving repetitive action potentials in neurons. The approach presented here offers a viable pathway toward customization of complex physiological properties of optogenetic tools. We propose that our screening method not only enables elucidation of new ChR variants that affect microbial opsin performance but may also reveal new principles of optogenetic protein engineering.
Collapse
Affiliation(s)
- Yong Ku Cho
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Demian Park
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Aimei Yang
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Fei Chen
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Amy S Chuong
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Nathan C Klapoetke
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| | - Edward S Boyden
- From the MIT Media Lab, McGovern Institute, and Koch Institute, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139
| |
Collapse
|
78
|
Serita T, Miyahara M, Tanimizu T, Takahashi S, Oishi S, Nagayoshi T, Tsuji R, Inoue H, Uehara M, Kida S. Dietary magnesium deficiency impairs hippocampus-dependent memories without changes in the spine density and morphology of hippocampal neurons in mice. Brain Res Bull 2019; 144:149-157. [DOI: 10.1016/j.brainresbull.2018.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 11/16/2018] [Accepted: 11/24/2018] [Indexed: 11/26/2022]
|
79
|
Cahill EN, Milton AL. Neurochemical and molecular mechanisms underlying the retrieval-extinction effect. Psychopharmacology (Berl) 2019; 236:111-132. [PMID: 30656364 PMCID: PMC6373198 DOI: 10.1007/s00213-018-5121-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/12/2018] [Indexed: 12/26/2022]
Abstract
Extinction within the reconsolidation window, or 'retrieval-extinction', has received much research interest as a possible technique for targeting the reconsolidation of maladaptive memories with a behavioural intervention. However, it remains to be determined whether the retrieval-extinction effect-a long-term reduction in fear behaviour, which appears resistant to spontaneous recovery, renewal and reinstatement-depends specifically on destabilisation of the original memory (the 'reconsolidation-update' account) or represents facilitation of an extinction memory (the 'extinction-facilitation' account). We propose that comparing the neurotransmitter systems, receptors and intracellular signalling pathways recruited by reconsolidation, extinction and retrieval-extinction will provide a way of distinguishing between these accounts.
Collapse
Affiliation(s)
- Emma N Cahill
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge, CB2 3EG, UK
| | - Amy L Milton
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK.
- Behavioural and Clinical Neuroscience Institute, Cambridge, CB2 3EB, UK.
| |
Collapse
|
80
|
Feldmann KG, Chowdhury A, Becker JL, McAlpin N, Ahmed T, Haider S, Richard Xia JX, Diaz K, Mehta MG, Mano I. Non-canonical activation of CREB mediates neuroprotection in a Caenorhabditis elegans model of excitotoxic necrosis. J Neurochem 2018; 148:531-549. [PMID: 30447010 DOI: 10.1111/jnc.14629] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/26/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
Excitotoxicity, caused by exaggerated neuronal stimulation by Glutamate (Glu), is a major cause of neurodegeneration in brain ischemia. While we know that neurodegeneration is triggered by overstimulation of Glu-receptors (GluRs), the subsequent mechanisms that lead to cellular demise remain controversial. Surprisingly, signaling downstream of GluRs can also activate neuroprotective pathways. The strongest evidence involves activation of the transcription factor cAMP response element-binding protein (CREB), widely recognized for its importance in synaptic plasticity. Canonical views describe CREB as a phosphorylation-triggered transcription factor, where transcriptional activation involves CREB phosphorylation and association with CREB-binding protein. However, given CREB's ubiquitous cross-tissue expression, the multitude of cascades leading to CREB phosphorylation, and its ability to regulate thousands of genes, it remains unclear how CREB exerts closely tailored, differential neuroprotective responses in excitotoxicity. A non-canonical, alternative cascade for activation of CREB-mediated transcription involves the CREB co-factor cAMP-regulated transcriptional co-activator (CRTC), and may be independent of CREB phosphorylation. To identify cascades that activate CREB in excitotoxicity we used a Caenorhabditis elegans model of neurodegeneration by excitotoxic necrosis. We demonstrated that CREB's neuroprotective effect was conserved, and seemed most effective in neurons with moderate Glu exposure. We found that factors mediating canonical CREB activation were not involved. Instead, phosphorylation-independent CREB activation in nematode excitotoxic necrosis hinged on CRTC. CREB-mediated transcription that depends on CRTC, but not on CREB phosphorylation, might lead to expression of a specific subset of neuroprotective genes. Elucidating conserved mechanisms of excitotoxicity-specific CREB activation can help us focus on core neuroprotective programs in excitotoxicity. Cover Image for this issue: doi: 10.1111/jnc.14494.
Collapse
Affiliation(s)
- K Genevieve Feldmann
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA
| | - Ayesha Chowdhury
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA
| | - Jessica L Becker
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - N'Gina McAlpin
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - Taqwa Ahmed
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Syed Haider
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - Jian X Richard Xia
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Karina Diaz
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Monal G Mehta
- Robert Wood Johnson Medical School, Rutgers - The State University of New Jersey, Piscataway, New Jersey, USA
| | - Itzhak Mano
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA.,The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| |
Collapse
|
81
|
Al-Attar R, Storey KB. Effects of anoxic exposure on the nuclear factor of activated T cell (NFAT) transcription factors in the stress-tolerant wood frog. Cell Biochem Funct 2018; 36:420-430. [PMID: 30411386 DOI: 10.1002/cbf.3362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/15/2018] [Accepted: 10/03/2018] [Indexed: 11/08/2022]
Abstract
The wood frog, Lithobates sylvaticus (also known as Rana sylvatica), is used for studying natural freeze tolerance. These animals convert 65% to 70% of their total body water into extracellular ice and survive freezing for weeks in winter. Freezing interrupts oxygen delivery to organs; thus, wood frogs limit their ATP usage by depressing their metabolism and redirecting the available energy only to prosurvival processes. Here, we studied the nuclear factor of activated T cell (NFAT) transcription factor family in response to 24-hour anoxia, and 4-hour aerobic recovery in liver and skeletal muscle. Protein expression levels of NFATc1-c4, calcineurin A and glycogen synthase kinase 3β (NFAT regulators), osteopontin, and atrial natriuretic peptide (ANP) (targets of NFATc3 and NFATc4, respectively) were measured by immunoblotting, and the DNA-binding activities of NFATc1-c4 were measured by DNA-protein interaction ELISAs. Results show that NFATc4, calcineurin, and ANP protein expression as well as NFATc4 DNA binding increased during anoxia in liver where calcineurin and ANP protein levels and NFATc4 DNA binding remaining high after aerobic recovery. Anoxia caused a significant increase in NFATc3 protein expression but not DNA-binding activity in muscle. Our results show that anoxia can increase NFATc4 transcriptional activity in liver, leading to the increase in expression of cytoprotective genes in the wood frog. Understanding the molecular mechanisms involved in mediating survival under anoxia/reoxygenation conditions in a naturally stress-tolerant model, such as the wood frog, provides insightful information on the prosurvival regulatory mechanisms involved in combating stress. This information will also further our understanding of metabolic rate depression and answer the question of how frogs tolerate prolonged periods of oxygen deprivation and resume to full function upon recovery without facing any detrimental side effects as other animals would.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Institude of Biochemistry and Department of Biology, Carleton University, Ottawa, Canada
| | - Kenneth B Storey
- Institude of Biochemistry and Department of Biology, Carleton University, Ottawa, Canada
| |
Collapse
|
82
|
Wheaton KL, Hansen KF, Aten S, Sullivan KA, Yoon H, Hoyt KR, Obrietan K. The Phosphorylation of CREB at Serine 133 Is a Key Event for Circadian Clock Timing and Entrainment in the Suprachiasmatic Nucleus. J Biol Rhythms 2018; 33:497-514. [PMID: 30175684 DOI: 10.1177/0748730418791713] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Within the suprachiasmatic nucleus (SCN)-the locus of the master circadian clock- transcriptional regulation via the CREB/CRE pathway is implicated in the functioning of the molecular clock timing process, and is a key conduit through which photic input entrains the oscillator. One event driving CRE-mediated transcription is the phosphorylation of CREB at serine 133 (Ser133). Indeed, numerous reporter gene assays have shown that an alanine point mutation in Ser133 reduces CREB-mediated transcription. Here, we sought to examine the contribution of Ser133 phosphorylation to the functional role of CREB in SCN clock physiology in vivo. To this end, we used a CREB knock-in mouse strain, in which Ser133 was mutated to alanine (S/A CREB). Under a standard 12 h light-dark cycle, S/A CREB mice exhibited a marked alteration in clock-regulated wheel running activity. Relative to WT mice, S/A CREB mice had highly fragmented bouts of locomotor activity during the night phase, elevated daytime activity, and a delayed phase angle of entrainment. Further, under free-running conditions, S/A CREB mice had a significantly longer tau than WT mice and reduced activity amplitude. In S/A CREB mice, light-evoked clock entrainment, using both Aschoff type 1 and 6 h "jet lag" paradigms, was markedly reduced relative to WT mice. S/A CREB mice exhibited attenuated transcriptional drive, as assessed by examining both clock-gated and light-evoked gene expression. Finally, SCN slice culture imaging detected a marked disruption in cellular clock phase synchrony following a phase-resetting stimulus in S/A CREB mice. Together, these data indicate that signaling through CREB phosphorylation at Ser133 is critical for the functional fidelity of both SCN timing and entrainment.
Collapse
Affiliation(s)
- Kelin L Wheaton
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH
| | | | - Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Kyle A Sullivan
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Hyojung Yoon
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH
| |
Collapse
|
83
|
Joshi RS, Panicker MM. Identifying the In Vivo Cellular Correlates of Antipsychotic Drugs. eNeuro 2018; 5:ENEURO.0220-18.2018. [PMID: 30713996 PMCID: PMC6354787 DOI: 10.1523/eneuro.0220-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/26/2018] [Accepted: 09/01/2018] [Indexed: 11/21/2022] Open
Abstract
GPCRs such as 5-HT2A and D2 are implicated in the therapeutic and the side effects of antipsychotics. However, the pattern of brain activity that leads to the behavioral effects of antipsychotics is poorly understood. To address this question, we used the transgenic 'FosTRAP' mice (Mus musculus), where a fluorescent reporter marks the cells responsive to the stimulus of interest. Here, the stimulus was an administration of various antipsychotic drugs. In case of typical antipsychotics such as Haloperidol, the c-fos active cells were predominantly found in the striatum, whereas in case of the atypical antipsychotics (Clozapine and Olanzapine), c-fos-induced cells were more numerous in the cortical regions, e.g., orbital cortex, piriform cortex. Curiously, we also observed ependymal cells to be a novel cellular target of atypical antipsychotics. 5-HT2A is considered to be a major target for atypical antipsychotics. Therefore, we bred 'FosTRAP' mice with 5-HT2A knock-out (KO) mice and tested their response to the prototype of atypical antipsychotics, Clozapine. Interestingly, the absence of 5-HT2A did not significantly affect the number of c-fos-induced cells in the cortical regions. However, the ependymal cells showed a dramatically reduced response to Clozapine in the absence of 5-HT2A. In summary, the TRAP system has allowed us to identify various region-specific activity induced by antipsychotics and novel cellular targets of the antipsychotics. These results serve as a "proof of principle" study that can be extended to explore the biochemical and physiological changes brought about by antipsychotics and specifically identify antipsychotic-responsive cells in the live tissue.
Collapse
Affiliation(s)
- Radhika S. Joshi
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bengaluru 560065, India
| | - Mitradas M. Panicker
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bengaluru 560065, India
| |
Collapse
|
84
|
β-Subunit of the voltage-gated Ca 2+ channel Cav1.2 drives signaling to the nucleus via H-Ras. Proc Natl Acad Sci U S A 2018; 115:E8624-E8633. [PMID: 30150369 DOI: 10.1073/pnas.1805380115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Depolarization-induced signaling to the nucleus by the L-type voltage-gated calcium channel Cav1.2 is widely assumed to proceed by elevating intracellular calcium. The apparent lack of quantitative correlation between Ca2+ influx and gene activation suggests an alternative activation pathway. Here, we demonstrate that membrane depolarization of HEK293 cells transfected with α11.2/β2b/α2δ subunits (Cav1.2) triggers c-Fos and MeCP2 activation via the Ras/ERK/CREB pathway. Nuclear signaling is lost either by absence of the intracellular β2 subunit or by transfecting the cells with the channel mutant α11.2W440A/β2b/α2δ, a mutation that disrupts the interaction between α11.2 and β2 subunits. Pulldown assays in neuronal SH-SY5Y cells and in vitro binding of recombinant H-Ras and β2 confirmed the importance of the intracellular β2 subunit for depolarization-induced gene activation. Using a Ca2+-impermeable mutant channel α11.2L745P/β2b/α2δ or disrupting Ca2+/calmodulin binding to the channel using the channel mutant α11.2I1624A/β2b/α2δ, we demonstrate that depolarization-induced c-Fos and MeCP2 activation does not depend on Ca2+ transport by the channel. Thus, in contrast to the paradigm that elevated intracellular Ca2+ drives nuclear signaling, we show that Cav1.2-triggered c-Fos or MeCP2 is dependent on extracellular Ca2+ and Ca2+ occupancy of the open channel pore, but is Ca2+-influx independent. An indispensable β-subunit interaction with H-Ras, which is triggered by conformational changes at α11.2 independently of Ca2+ flux, brings to light a master regulatory role of β2 in transcriptional activation via the ERK/CREB pathway. This mode of H-Ras activation could have broad implications for understanding the coupling of membrane depolarization to the rapid induction of gene transcription.
Collapse
|
85
|
A novel environment-evoked transcriptional signature predicts reactivity in single dentate granule neurons. Nat Commun 2018; 9:3084. [PMID: 30082781 PMCID: PMC6079101 DOI: 10.1038/s41467-018-05418-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Activity-induced remodeling of neuronal circuits is critical for memory formation. This process relies in part on transcription, but neither the rate of activity nor baseline transcription is equal across neuronal cell types. In this study, we isolated mouse hippocampal populations with different activity levels and used single nucleus RNA-seq to compare their transcriptional responses to activation. One hour after novel environment exposure, sparsely active dentate granule (DG) neurons had a much stronger transcriptional response compared to more highly active CA1 pyramidal cells and vasoactive intestinal polypeptide (VIP) interneurons. Activity continued to impact transcription in DG neurons up to 5 h, with increased heterogeneity. By re-exposing the mice to the same environment, we identified a unique transcriptional signature that selects DG neurons for reactivation upon re-exposure to the same environment. These results link transcriptional heterogeneity to functional heterogeneity and identify a transcriptional correlate of memory encoding in individual DG neurons. Single nuclei RNA-seq has been used to characterize transcriptional signature of environment-related activity in cells of the dentate gyrus. Here the authors use this approach to show that whether a neuron will be reactivated in response to re-exposure to a previous environment can be predicted by its transcriptional signature.
Collapse
|
86
|
Olfactory-Experience- and Developmental-Stage-Dependent Control of CPEB4 Regulates c-Fos mRNA Translation for Granule Cell Survival. Cell Rep 2018; 21:2264-2276. [PMID: 29166615 DOI: 10.1016/j.celrep.2017.10.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/13/2017] [Accepted: 10/25/2017] [Indexed: 11/21/2022] Open
Abstract
Mammalian olfactory bulbs (OBs) require continuous replenishment of interneurons (mainly granule cells [GCs]) to support local circuits throughout life. Two spatiotemporally distinct waves of postnatal neurogenesis contribute to expanding and maintaining the GC pool. Although neonate-born GCs have a higher survival rate than adult-born GCs, the molecular mechanism underlying this survival remains unclear. Here, we find that cytoplasmic polyadenylation element-binding protein 4 (CPEB4) acts as a survival factor exclusively for early postnatal GCs. In mice, during the first 2 postnatal weeks, olfactory experience initiated CPEB4-activated c-Fos mRNA translation. In CPEB4-knockout mice, c-FOS insufficiency reduced neurotrophic signaling to impair GC survival and cause OB hypoplasia. Both cyclic AMP responsive element binding protein (CREB)-dependent transcription and CPEB4-promoted translation support c-FOS expression early postnatal OBs but disengage in adult OBs. Activity-related c-FOS synthesis and GC survival are thus developmentally controlled by distinct molecular mechanisms to govern OB growth.
Collapse
|
87
|
Policarpi C, Crepaldi L, Brookes E, Nitarska J, French SM, Coatti A, Riccio A. Enhancer SINEs Link Pol III to Pol II Transcription in Neurons. Cell Rep 2018; 21:2879-2894. [PMID: 29212033 PMCID: PMC5732322 DOI: 10.1016/j.celrep.2017.11.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 06/30/2017] [Accepted: 11/02/2017] [Indexed: 12/25/2022] Open
Abstract
Spatiotemporal regulation of gene expression depends on the cooperation of multiple mechanisms, including the functional interaction of promoters with distally located enhancers. Here, we show that, in cortical neurons, a subset of short interspersed nuclear elements (SINEs) located in the proximity of activity-regulated genes bears features of enhancers. Enhancer SINEs (eSINEs) recruit the Pol III cofactor complex TFIIIC in a stimulus-dependent manner and are transcribed by Pol III in response to neuronal depolarization. Characterization of an eSINE located in proximity to the Fos gene (FosRSINE1) indicated that the FosRSINE1-encoded transcript interacts with Pol II at the Fos promoter and mediates Fos relocation to Pol II factories, providing an unprecedented molecular link between Pol III and Pol II transcription. Strikingly, knockdown of the FosRSINE1 transcript induces defects of both cortical radial migration in vivo and activity-dependent dendritogenesis in vitro, demonstrating that FosRSINE1 acts as a strong enhancer of Fos expression in diverse physiological contexts.
Collapse
Affiliation(s)
- Cristina Policarpi
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Luca Crepaldi
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Emily Brookes
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Justyna Nitarska
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Sarah M French
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Alessandro Coatti
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Antonella Riccio
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
88
|
Breit M, Queisser G. What Is Required for Neuronal Calcium Waves? A Numerical Parameter Study. JOURNAL OF MATHEMATICAL NEUROSCIENCE 2018; 8:9. [PMID: 30006849 PMCID: PMC6045568 DOI: 10.1186/s13408-018-0064-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/15/2018] [Indexed: 06/03/2023]
Abstract
Neuronal calcium signals propagating by simple diffusion and reaction with mobile and stationary buffers are limited to cellular microdomains. The distance intracellular calcium signals can travel may be significantly increased by means of calcium-induced calcium release from internal calcium stores, notably the endoplasmic reticulum. The organelle, which can be thought of as a cell-within-a-cell, is able to sequester large amounts of cytosolic calcium ions via SERCA pumps and selectively release them into the cytosol through ryanodine receptor channels leading to the formation of calcium waves. In this study, we set out to investigate the basic properties of such dendritic calcium waves and how they depend on the three parameters dendrite radius, ER radius and ryanodine receptor density in the endoplasmic membrane. We demonstrate that there are stable and abortive regimes for calcium waves, depending on the above morphological and physiological parameters. In stable regimes, calcium waves can travel across long dendritic distances, similar to electrical action potentials. We further observe that abortive regimes exist, which could be relevant for spike-timing dependent plasticity, as travel distances and wave velocities vary with changing intracellular architecture. For some of these regimes, analytic functions could be derived that fit the simulation data. In parameter spaces, that are non-trivially influenced by the three-dimensional calcium concentration profile, we were not able to derive such a functional description, demonstrating the mathematical requirement to model and simulate biochemical signaling in three-dimensional space.
Collapse
Affiliation(s)
- Markus Breit
- G-CSC, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
89
|
Alfonso-Gonzalez C, Riesgo-Escovar JR. Fos metamorphoses: Lessons from mutants in model organisms. Mech Dev 2018; 154:73-81. [PMID: 29753813 DOI: 10.1016/j.mod.2018.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
The Fos oncogene gene family is evolutionarily conserved throughout Eukarya. Fos proteins characteristically have a leucine zipper and a basic region with a helix-turn-helix motif that binds DNA. In vertebrates, there are several Fos homologs. They can homo- or hetero-dimerize via the leucine zipper domain. Fos homologs coupled with other transcription factors, like Jun oncoproteins, constitute the Activator Protein 1 (AP-1) complex. From its original inception as an oncogene, the subsequent finding that they act as transcription factors binding DNA sequences known as TRE, to the realization that they are activated in many different scenarios, and to loss-of-function analysis, the Fos proteins have traversed a multifarious path in development and physiology. They are instrumental in 'immediate early genes' responses, and activated by a seemingly myriad assemblage of different stimuli. Yet, the majority of these studies were basically gain-of-function studies, since it was thought that Fos genes would be cell lethal. Loss-of-function mutations in vertebrates were recovered later, and were not cell lethal. In fact, c-fos null mutations are viable with developmental defects (osteopetrosis and myeloid lineage abnormalities). It was then hypothesized that vertebrate genomes exhibit partial redundancy, explaining the 'mild' phenotypes, and complicating assessment of complete loss-of-function phenotypes. Due to its promiscuous activation, fos genes (especially c-fos) are now commonly used as markers for cellular responses to stimuli. fos homologs high sequence conservation (including Drosophila) is advantageous as it allows critical assessment of fos genes functions in this genetic model. Drosophila melanogaster contains only one fos homolog, the gene kayak. kayak mutations are lethal, and allow study of all the processes where fos is required. The kayak locus encodes several different isoforms, and is a pleiotropic gene variously required for development involving cell shape changes. In general, fos genes seem to primarily activate programs involved in cellular architectural rearrangements and cell shape changes.
Collapse
Affiliation(s)
- Carlos Alfonso-Gonzalez
- Developmental Neurobiology and Neurophysiology Department, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro c.p.76230, Mexico; Maestría en Bioquímica y Biología Molecular, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Juan Rafael Riesgo-Escovar
- Developmental Neurobiology and Neurophysiology Department, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro c.p.76230, Mexico.
| |
Collapse
|
90
|
What does the Fos say? Using Fos-based approaches to understand the contribution of stress to substance use disorders. Neurobiol Stress 2018; 9:271-285. [PMID: 30450391 PMCID: PMC6234265 DOI: 10.1016/j.ynstr.2018.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/08/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023] Open
Abstract
Despite extensive research efforts, drug addiction persists as a largely unmet medical need. Perhaps the biggest challenge for treating addiction is the high rate of recidivism. While many factors can promote relapse in abstinent drug users, the contribution of stress is particularly problematic, as stress is uncontrollable and pervasive in the lives of those struggling with addiction. Thus, understanding the neurocircuitry that underlies the influence of stress on drug seeking is critical for guiding treatment. Preclinical research aimed at defining this neurocircuitry has, in part, relied upon the use of experimental approaches that allow visualization of cellular and circuit activity that corresponds to stressor-induced drug seeking in rodent relapse models. Much of what we have learned about the mechanisms that mediate stressor-induced relapse has been informed by studies that have used the expression of the immediate early gene, cfos, or its protein product, Fos, as post-mortem activity markers. In this review we provide an overview of the rodent models used to study stressor-induced relapse and briefly summarize what is known about the underlying neurocircuitry before describing the use of cfos/Fos-based approaches. In addition to reviewing findings obtained using this approach, its advantages and limitations are considered. Moreover, new techniques that leverage the expression profile of cfos to tag and manipulate cells based on their activity patterns are discussed. The intent of the review is to guide the interpretation of old and design of new studies that utilize cfos/Fos-based strategies to study the neurocircuitry that contributes to stress-related drug use.
Collapse
|
91
|
Das S, Moon HC, Singer RH, Park HY. A transgenic mouse for imaging activity-dependent dynamics of endogenous Arc mRNA in live neurons. SCIENCE ADVANCES 2018; 4:eaar3448. [PMID: 29938222 PMCID: PMC6010337 DOI: 10.1126/sciadv.aar3448] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/10/2018] [Indexed: 05/05/2023]
Abstract
Localized translation plays a crucial role in synaptic plasticity and memory consolidation. However, it has not been possible to follow the dynamics of memory-associated mRNAs in living neurons in response to neuronal activity in real time. We have generated a novel mouse model where the endogenous Arc/Arg3.1 gene is tagged in its 3' untranslated region with stem-loops that bind a bacteriophage PP7 coat protein (PCP), allowing visualization of individual mRNAs in real time. The physiological response of the tagged gene to neuronal activity is identical to endogenous Arc and reports the true dynamics of Arc mRNA from transcription to degradation. The transcription dynamics of Arc in cultured hippocampal neurons revealed two novel results: (i) A robust transcriptional burst with prolonged ON state occurs after stimulation, and (ii) transcription cycles continue even after initial stimulation is removed. The correlation of stimulation with Arc transcription and mRNA transport in individual neurons revealed that stimulus-induced Ca2+ activity was necessary but not sufficient for triggering Arc transcription and that blocking neuronal activity did not affect the dendritic transport of newly synthesized Arc mRNAs. This mouse will provide an important reagent to investigate how individual neurons transduce activity into spatiotemporal regulation of gene expression at the synapse.
Collapse
Affiliation(s)
- Sulagna Das
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hyungseok C. Moon
- Department of Physics and Astronomy, Seoul National University, Seoul 08826, Korea
| | - Robert H. Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
- Corresponding author. (H.Y.P.); (R.H.S.)
| | - Hye Yoon Park
- Department of Physics and Astronomy, Seoul National University, Seoul 08826, Korea
- The Institute of Applied Physics, Seoul National University, Seoul 08826, Korea
- Corresponding author. (H.Y.P.); (R.H.S.)
| |
Collapse
|
92
|
Thiel G, Rössler OG. Resveratrol stimulates c-Fos gene transcription via activation of ERK1/2 involving multiple genetic elements. Gene 2018. [PMID: 29514046 DOI: 10.1016/j.gene.2018.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The polyphenol resveratrol is found in many plant and fruits and is a constituent of our diet. Resveratrol has been proposed to have chemopreventive and anti-inflammatory activities. On the cellular level, resveratrol activates stimulus-regulated transcription factors. To identify resveratrol-responsive elements within a natural gene promoter, the molecular pathway leading to c-Fos gene expression by resveratrol was dissected. The c-Fos gene encodes a basic region leucine zipper transcription factor and is a prototype of an immediate-early gene that is regulated by a wide range of signaling molecules. We analyzed chromatin-integrated c-Fos promoter-luciferase reporter genes where transcription factor binding sites were destroyed by point mutations or deletion mutagenesis. The results show that mutation of the binding sites for serum response factor (SRF), activator protein-1 (AP-1) and cAMP response element binding protein (CREB) significantly reduced reporter gene transcription following stimulation of the cells with resveratrol. Inactivation of the binding sites for signal transducer and activator of transcription (STAT) or ternary complex factors did not influence resveratrol-regulated c-Fos promoter activity. Thus, the c-Fos promoter contains three resveratrol-responsive elements, the cAMP response element (CRE), and the binding sites for SRF and AP-1. Moreover, we show that the transcriptional activation potential of the c-Fos protein is increased in resveratrol-stimulated cells, indicating that the biological activity of c-Fos is elevated by resveratrol stimulation. Pharmacological and genetic experiments revealed that the protein kinase ERK1/2 is the signal transducer that connects resveratrol treatment with the c-Fos gene.
Collapse
Affiliation(s)
- Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany.
| | - Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| |
Collapse
|
93
|
Zimbone S, Monaco I, Gianì F, Pandini G, Copani AG, Giuffrida ML, Rizzarelli E. Amyloid Beta monomers regulate cyclic adenosine monophosphate response element binding protein functions by activating type-1 insulin-like growth factor receptors in neuronal cells. Aging Cell 2018; 17. [PMID: 29094448 PMCID: PMC5770784 DOI: 10.1111/acel.12684] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2017] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with synaptic dysfunction, pathological accumulation of β-amyloid (Aβ), and neuronal loss. The self-association of Aβ monomers into soluble oligomers seems to be crucial for the development of neurotoxicity (J. Neurochem., 00, 2007 and 1172). Aβ oligomers have been suggested to compromise neuronal functions in AD by reducing the expression levels of the CREB target gene and brain-derived neurotrophic factor (BDNF) (J. Neurosci., 27, 2007 and 2628; Neurobiol. Aging, 36, 2015 and 20406 Mol. Neurodegener., 6, 2011 and 60). We previously reported a broad neuroprotective activity of physiological Aβ monomers, involving the activation of type-1 insulin-like growth factor receptors (IGF-IRs) (J. Neurosci., 29, 2009 and 10582, Front Cell Neurosci., 9, 2015 and 297). We now provide evidence that Aβ monomers, by activating the IGF-IR-stimulated PI3-K/AKT pathway, induce the activation of CREB in neurons and sustain BDNF transcription and release.
Collapse
Affiliation(s)
- Stefania Zimbone
- Institute of Biostructures and Bioimaging; National Council of Research (IBB-CNR); Via Paolo Gaifami 18 95126 Catania Italy
| | - Irene Monaco
- Institute of Biostructures and Bioimaging; National Council of Research (IBB-CNR); Via Paolo Gaifami 18 95126 Catania Italy
| | - Fiorenza Gianì
- Endocrinology, Department of Clinical and Experimental Medicine; Garibaldi-Nesima Medical Center; University of Catania; via Palermo 636 95122 Catania Italy
| | - Giuseppe Pandini
- Endocrinology, Department of Clinical and Experimental Medicine; Garibaldi-Nesima Medical Center; University of Catania; via Palermo 636 95122 Catania Italy
| | - Agata G. Copani
- Institute of Biostructures and Bioimaging; National Council of Research (IBB-CNR); Via Paolo Gaifami 18 95126 Catania Italy
- Department of Drug Sciences; University of Catania; Viale A. Doria 6 95125 Catania Italy
| | - Maria Laura Giuffrida
- Institute of Biostructures and Bioimaging; National Council of Research (IBB-CNR); Via Paolo Gaifami 18 95126 Catania Italy
| | - Enrico Rizzarelli
- Institute of Biostructures and Bioimaging; National Council of Research (IBB-CNR); Via Paolo Gaifami 18 95126 Catania Italy
- Department of Chemical Sciences; University of Catania; Viale A. Doria 6 95125 Catania Italy
| |
Collapse
|
94
|
Lee HY, Itahana Y, Schuechner S, Fukuda M, Je HS, Ogris E, Virshup DM, Itahana K. Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation–induced cell death independently of inhibiting glycolysis. Sci Signal 2018; 11:11/512/eaam7893. [DOI: 10.1126/scisignal.aam7893] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
95
|
POU6f1 Mediates Neuropeptide-Dependent Plasticity in the Adult Brain. J Neurosci 2018; 38:1443-1461. [PMID: 29305536 DOI: 10.1523/jneurosci.1641-17.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/11/2017] [Accepted: 12/21/2017] [Indexed: 01/20/2023] Open
Abstract
The mouse olfactory bulb (OB) features continued, activity-dependent integration of adult-born neurons, providing a robust model with which to examine mechanisms of plasticity in the adult brain. We previously reported that local OB interneurons secrete the neuropeptide corticotropin-releasing hormone (CRH) in an activity-dependent manner onto adult-born granule neurons and that local CRH signaling promotes expression of synaptic machinery in the bulb. This effect is mediated via activation of the CRH receptor 1 (CRHR1), which is developmentally regulated during adult-born neuron maturation. CRHR1 is a GS-protein-coupled receptor that activates CREB-dependent transcription in the presence of CRH. Therefore, we hypothesized that locally secreted CRH activates CRHR1 to initiate circuit plasticity programs. To identify such programs, we profiled gene expression changes associated with CRHR1 activity in adult-born neurons of the OB. Here, we show that CRHR1 activity influences expression of the brain-specific Homeobox-containing transcription factor POU Class 6 Homeobox 1 (POU6f1). To elucidate the contributions of POU6f1 toward activity-dependent circuit remodeling, we targeted CRHR1+ neurons in male and female mice for cell-type-specific manipulation of POU6f1 expression. Whereas loss of POU6f1 in CRHR1+ neurons resulted in reduced dendritic complexity and decreased synaptic connectivity, overexpression of POU6f1 in CRHR1+ neurons promoted dendritic outgrowth and branching and influenced synaptic function. Together, these findings suggest that the transcriptional program directed by POU6f1 downstream of local CRH signaling in adult-born neurons influences circuit dynamics in response to activity-dependent peptide signaling in the adult brain.SIGNIFICANCE STATEMENT Elucidating mechanisms of plasticity in the adult brain is helpful for devising strategies to understand and treat neurodegeneration. Circuit plasticity in the adult mouse olfactory bulb is exemplified by both continued cell integration and synaptogenesis. We previously reported that these processes are influenced by local neuropeptide signaling in an activity-dependent manner. Here, we show that local corticotropin-releasing hormone (CRH) signaling induces dynamic gene expression changes in CRH receptor expressing adult-born neurons, including altered expression of the transcription factor POU6f1 We further show that POU6f1 is necessary for proper dendrite specification and patterning, as well as synapse development and function in adult-born neurons. Together, these findings reveal a novel mechanism by which peptide signaling modulates adult brain circuit plasticity.
Collapse
|
96
|
Eagle AL, Gajewski PA, Robison AJ. Role of hippocampal activity-induced transcription in memory consolidation. Rev Neurosci 2018; 27:559-73. [PMID: 27180338 DOI: 10.1515/revneuro-2016-0010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/26/2016] [Indexed: 01/15/2023]
Abstract
Experience-dependent changes in the strength of connections between neurons in the hippocampus (HPC) are critical for normal learning and memory consolidation, and disruption of this process drives a variety of neurological and psychiatric diseases. Proper HPC function relies upon discrete changes in gene expression driven by transcription factors (TFs) induced by neuronal activity. Here, we describe the induction and function of many of the most well-studied HPC TFs, including cyclic-AMP response element binding protein, serum-response factor, AP-1, and others, and describe their role in the learning process. We also discuss the known target genes of many of these TFs and the purported mechanisms by which they regulate long-term changes in HPC synaptic strength. Moreover, we propose that future research in this field will depend upon unbiased identification of additional gene targets for these activity-dependent TFs and subsequent meta-analyses that identify common genes or pathways regulated by multiple TFs in the HPC during learning or disease.
Collapse
|
97
|
Dolzani SD, Baratta MV, Moss JM, Leslie NL, Tilden SG, Sørensen AT, Watkins LR, Lin Y, Maier SF. Inhibition of a Descending Prefrontal Circuit Prevents Ketamine-Induced Stress Resilience in Females. eNeuro 2018; 5:ENEURO.0025-18.2018. [PMID: 29516036 PMCID: PMC5839773 DOI: 10.1523/eneuro.0025-18.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/23/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022] Open
Abstract
Stress is a potent etiological factor in the onset of major depressive disorder and posttraumatic stress disorder (PTSD). Therefore, significant efforts have been made to identify factors that produce resilience to the outcomes of a later stressor, in hopes of preventing untoward clinical outcomes. The NMDA receptor antagonist ketamine has recently emerged as a prophylactic capable of preventing neurochemical and behavioral outcomes of a future stressor. Despite promising results of preclinical studies performed in male rats, the effects of proactive ketamine in female rats remains unknown. This is alarming given that stress-related disorders affect females at nearly twice the rate of males. Here we explore the prophylactic effects of ketamine on stress-induced anxiety-like behavior and the neural circuit-level processes that mediate these effects in female rats. Ketamine given one week prior to an uncontrollable stressor (inescapable tailshock; IS) reduced typical stress-induced activation of the serotonergic (5-HT) dorsal raphe nucleus (DRN) and eliminated DRN-dependent juvenile social exploration (JSE) deficits 24 h after the stressor. Proactive ketamine altered prelimbic cortex (PL) neural ensembles so that a later experience with IS now activated these cells, which it ordinarily would not. Ketamine acutely activated a PL to DRN (PL-DRN) circuit and inhibition of this circuit with Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) at the time of IS one week later prevented stress prophylaxis, suggesting that persistent changes in PL-DRN circuit activity are responsible, at least in part, for mediating long-term effects associated with ketamine.
Collapse
Affiliation(s)
- S D Dolzani
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80309
| | - M V Baratta
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| | - J M Moss
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| | - N L Leslie
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| | - S G Tilden
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| | - A T Sørensen
- Department of Neuroscience, University of Copenhagen, Copenhagen, 1165 Denmark
| | - L R Watkins
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| | - Y Lin
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - S F Maier
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309
| |
Collapse
|
98
|
Jaworski J, Kalita K, Knapska E. c-Fos and neuronal plasticity: the aftermath of Kaczmarek’s theory. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
99
|
CRTC1 mediates preferential transcription at neuronal activity-regulated CRE/TATA promoters. Sci Rep 2017; 7:18004. [PMID: 29269871 PMCID: PMC5740062 DOI: 10.1038/s41598-017-18215-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/27/2017] [Indexed: 01/03/2023] Open
Abstract
Gene expression mediated by the transcription factor cAMP-responsive element-binding protein (CREB) is essential for a wide range of brain processes. The transcriptional coactivartor CREB-regulated transcription coactivator-1 (CRTC1) is required for efficient induction of CREB target genes during neuronal activity. However, the mechanisms regulating induction of specific CREB/CRTC1-dependent genes during neuronal activity remain largely unclear. Here, we investigated the molecular mechanisms regulating activity-dependent gene transcription upon activation of the CREB/CRTC1 signaling pathway in neurons. Depolarization and cAMP signals induce preferential transcription of activity-dependent genes containing promoters with proximal CRE/TATA sequences, such as c-fos, Dusp1, Nr4a1, Nr4a2 and Ptgs2, but not genes with proximal CRE/TATA-less promoters (e.g. Nr4a3, Presenilin-1 and Presenilin-2). Notably, biochemical and chromatin immunoprecipitation analyses reveal constitutive binding of CREB to target gene promoters in the absence of neuronal activity, whereas recruitment of CRTC1 to proximal CRE/TATA promoters depends on neuronal activity. Neuronal activity induces rapid CRTC1 dephosphorylation, nuclear translocation and binding to endogenous CREB. These results indicate that neuronal activity induces a preferential binding of CRTC1 to the transcriptional complex in CRE/TATA-containing promoters to engage activity-dependent transcription in neurons.
Collapse
|
100
|
Carter SD, Mifsud KR, Reul JMHM. Acute Stress Enhances Epigenetic Modifications But Does Not Affect the Constitutive Binding of pCREB to Immediate-Early Gene Promoters in the Rat Hippocampus. Front Mol Neurosci 2017; 10:416. [PMID: 29311809 PMCID: PMC5742222 DOI: 10.3389/fnmol.2017.00416] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/30/2017] [Indexed: 01/13/2023] Open
Abstract
The immediate early genes (IEGs) c-Fos and Egr-1 are rapidly and transiently induced in sparse neurons within the hippocampus after exposure to an acute stressor. The induction of these genes is a critical part of the molecular mechanisms underlying successful behavioral adaptation to stress. Our previous work has shown that transcriptional activation of c-Fos and Egr-1 in the hippocampus requires formation of a dual histone mark within their promoter regions, the phosphorylation of serine 10 and acetylation of lysine 9/14 of histone H3. In the present study, using chromatin immuno-precipitation (ChIP), we found that an increase in the formation of H3K9ac-S10p occurs within the c-Fos and Egr-1 promoters after FS stress in vivo and that these histone modifications were located to promoter regions containing cAMP Responsive Elements (CREs), but not in neighboring regions containing only Serum Responsive Elements (SREs). Surprisingly, however, subsequent ChIP analyses showed no changes in the binding of pCREB or CREB-binding protein (CBP) to the CREs after FS. In fact, pCREB binding to the c-Fos and Egr-1 promoters was already highly enriched under baseline conditions and did not increase further after stress. We suggest that constitutive pCREB binding may keep c-Fos and Egr-1 in a poised state for activation. Possibly, the formation of H3K9ac-S10p in the vicinity of CRE sites may participate in unblocking transcriptional elongation through recruitment of additional epigenetic factors.
Collapse
Affiliation(s)
| | | | - Johannes M. H. M. Reul
- Neuro-Epigenetics Research Group, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|