1
|
Foran G, Hallam RD, Megaly M, Turgambayeva A, Antfolk D, Li Y, Luca VC, Necakov A. Notch1 Phase Separation Coupled Percolation facilitates target gene expression and enhancer looping. Sci Rep 2024; 14:21912. [PMID: 39300145 PMCID: PMC11413390 DOI: 10.1038/s41598-024-71634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
The Notch receptor is a pleiotropic signaling protein that translates intercellular ligand interactions into changes in gene expression via the nuclear localization of the Notch intracellular Domain (NICD). Using a combination of immunohistochemistry, RNA in situ, Optogenetics and super-resolution live imaging of transcription in human cells, we show that the N1ICD can form condensates that positively facilitate Notch target gene expression. We determined that N1ICD undergoes Phase Separation Coupled Percolation (PSCP) into transcriptional condensates, which recruit, enrich, and encapsulate a broad set of core transcriptional proteins. We show that the capacity for condensation is due to the intrinsically disordered transcriptional activation domain of the N1ICD. In addition, the formation of such transcriptional condensates acts to promote Notch-mediated super enhancer-looping and concomitant activation of the MYC protooncogene expression. Overall, we introduce a novel mechanism of Notch1 activity in which discrete changes in nuclear N1ICD abundance are translated into the assembly of transcriptional condensates that facilitate gene expression by enriching essential transcriptional machineries at target genomic loci.
Collapse
Affiliation(s)
- Gregory Foran
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada
| | - Ryan Douglas Hallam
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada
| | - Marvel Megaly
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada
| | - Anel Turgambayeva
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada
| | - Daniel Antfolk
- Department of Immunology, Moffitt Cancer Centre, Tampa, FL, USA
| | - Yifeng Li
- Department of Computer Science, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada
| | - Vincent C Luca
- Department of Immunology, Moffitt Cancer Centre, Tampa, FL, USA
| | - Aleksandar Necakov
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
2
|
Foran G, Hallam RD, Megaly M, Turgambayeva A, Antfolk D, Li Y, Luca VC, Necakov A. Notch1 Phase Separation Coupled Percolation facilitates target gene expression and enhancer looping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.17.533124. [PMID: 39131356 PMCID: PMC11312450 DOI: 10.1101/2023.03.17.533124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The Notch receptor is a pleiotropic signaling protein that translates intercellular ligand interactions into changes in gene expression via the nuclear localization of the Notch intracellular Domain (NICD). Using a combination of immunohistochemistry, RNA in situ, Optogenetics and super-resolution live imaging of transcription in human cells, we show that the N1ICD can form condensates that positively facilitate Notch target gene expression. We determined that N1ICD undergoes Phase Separation Coupled Percolation (PSCP) into transcriptional condensates, which recruit, enrich, and encapsulate a broad set of core transcriptional proteins. We show that the capacity for condensation is due to the intrinsically disordered transcriptional activation domain of the N1ICD. In addition, the formation of such transcriptional condensates acts to promote Notch-mediated super enhancer-looping and concomitant activation of the MYC protooncogene expression. Overall, we introduce a novel mechanism of Notch1 activity in which discrete changes in nuclear N1ICD abundance are translated into the assembly of transcriptional condensates that facilitate gene expression by enriching essential transcriptional machineries at target genomic loci.
Collapse
Affiliation(s)
- Gregory Foran
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Ryan Douglas Hallam
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Marvel Megaly
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Anel Turgambayeva
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Daniel Antfolk
- Department of Immunology, Moffitt Cancer Centre, Tampa, FL, USA
| | - Yifeng Li
- Department of Computer Science, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Vincent C. Luca
- Department of Immunology, Moffitt Cancer Centre, Tampa, FL, USA
| | - Aleksandar Necakov
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| |
Collapse
|
3
|
Kim J, Kaang BK. Cyclic AMP response element-binding protein (CREB) transcription factor in astrocytic synaptic communication. Front Synaptic Neurosci 2023; 14:1059918. [PMID: 36685081 PMCID: PMC9845270 DOI: 10.3389/fnsyn.2022.1059918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/24/2022] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are known to actively participate in synaptic communication by forming structures called tripartite synapses. These synapses consist of presynaptic axon terminals, postsynaptic dendritic spines, and astrocytic processes where astrocytes release and receive transmitters. Although the transcription factor cyclic AMP response element (CRE)-binding protein (CREB) has been actively studied as an important factor for mediating synaptic activity-induced responses in neurons, its role in astrocytes is relatively unknown. Synaptic signals are known to activate various downstream pathways in astrocytes, which can activate the CREB transcription factor. Therefore, there is a need to summarize studies on astrocytic intracellular pathways that are induced by synaptic communication resulting in activation of the CREB pathway. In this review, we discuss the various neurotransmitter receptors and intracellular pathways that can induce CREB activation and CREB-induced gene regulation in astrocytes.
Collapse
|
4
|
Im DS, Joselin A, Svoboda D, Takano T, Rousseaux MWC, Callaghan S, Slack RS, Hisanaga SI, Davis RJ, Park DS, Qu D. Cdk5-mediated JIP1 phosphorylation regulates axonal outgrowth through Notch1 inhibition. BMC Biol 2022; 20:115. [PMID: 35581583 PMCID: PMC9115922 DOI: 10.1186/s12915-022-01312-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Activated Cdk5 regulates a number of processes during nervous system formation, including neuronal differentiation, growth cone stabilization, and axonal growth. Cdk5 phosphorylates its downstream substrates located in axonal growth cones, where the highly expressed c-Jun N-terminal kinase (JNK)-interacting protein1 (JIP1) has been implicated as another important regulator of axonal growth. In addition, stringent control of the level of intracellular domain of Notch1 (Notch1-IC) plays a regulatory role in axonal outgrowth during neuronal differentiation. However, whether Cdk5-JIP1-Notch1 cooperate to regulate axonal outgrowth, and the mechanism of such joint contribution to this pathway, is presently unknown, and here we explore their potential interaction. RESULTS Our interactome screen identified JIP1 as an interactor of p35, a Cdk5 activator, and we sought to explore the relationship between Cdk5 and JIP1 on the regulation of axonal outgrowth. We demonstrate that JIP1 phosphorylated by Cdk5 at Thr205 enhances axonal outgrowth and a phosphomimic JIP1 rescues the axonal outgrowth defects in JIP1-/- and p35-/- neurons. Axonal outgrowth defects caused by the specific increase of Notch1 in JIP1-/- neurons are rescued by Numb-mediated inhibition of Notch1. Finally, we demonstrate that Cdk5 phosphorylation of JIP1 further amplifies the phosphorylation status of yet another Cdk5 substrate E3-ubiquitin ligase Itch, resulting in increased Notch1 ubiquitination. CONCLUSIONS Our findings identify a potentially critical signaling axis involving Cdk5-JIP1-Itch-Notch1, which plays an important role in the regulation of CNS development. Future investigation into the way this pathway integrates with additional pathways regulating axonal growth will further our knowledge of normal central nervous system development and pathological conditions.
Collapse
Affiliation(s)
- Doo Soon Im
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Alvin Joselin
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Devon Svoboda
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Tesuya Takano
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Maxime W C Rousseaux
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Steve Callaghan
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01650, USA
| | - David S Park
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Dianbo Qu
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
5
|
Yao T, Xu Z, Hao Z, Yu Y, Liang B, Wang S. KDM5B promotes cell migration by regulating the noncanonical Wnt/PCP pathway in Hirschsprung's disease. Pediatr Surg Int 2022; 38:99-107. [PMID: 34455465 DOI: 10.1007/s00383-021-05005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE We measured the expression of the histone demethylase lysine-specific demethylase 5B (KDM5B) in the bowels of patients with Hirschsprung's disease (HSCR) and investigated the molecular mechanism by which KDM5B promotes the migration of neuronal PC12 cells. METHODS KDM5B expression was detected in the ganglionic and aganglionic colon of patients with HSCR (n = 10) and controls (n = 10). The expression and localization of KDM5B were assessed using immunohistochemical and immunofluorescence staining. Real-time PCR and Western blotting were performed to quantify KDM5B expression. The migration was determined using Transwell and wound-healing assays. G-LISA, GTPase pulldown and luciferase-based reporter gene assays were performed to evaluate the key components of Wnt/planar cell polarity (PCP) signaling in vitro. RESULTS Our current study showed that KDM5B colocalized with neurons. KDM5B expression was reduced in HSCR specimens, while the aganglionic segments showed the greatest reduction. KDM5B knockdown inhibited the migration of PC12 cells. Moreover, inhibition of KDM5B decreased the expression of key genes in the Wnt/PCP pathway, and its inhibitory effect on PC12 cell migration was reversed by Wnt5a treatment. CONCLUSIONS KDM5B promotes neuronal migration via the Wnt/PCP pathway. A potential role for KDM5B in altered enteric nervous system development in HSCR warrants further investigation.
Collapse
Affiliation(s)
- Ting Yao
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Zhilin Xu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Zenghui Hao
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - You Yu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Bingxue Liang
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Shuyu Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
6
|
Deyts C, Clutter M, Pierce N, Chakrabarty P, Ladd TB, Goddi A, Rosario AM, Cruz P, Vetrivel K, Wagner SL, Thinakaran G, Golde TE, Parent AT. APP-Mediated Signaling Prevents Memory Decline in Alzheimer's Disease Mouse Model. Cell Rep 2020; 27:1345-1355.e6. [PMID: 31042463 DOI: 10.1016/j.celrep.2019.03.087] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023] Open
Abstract
Amyloid precursor protein (APP) and its metabolites play key roles in Alzheimer's disease (AD) pathophysiology. Whereas short amyloid-β (Aβ) peptides derived from APP are pathogenic, the APP holoprotein serves multiple purposes in the nervous system through its cell adhesion and receptor-like properties. Our studies focused on the signaling mediated by the APP cytoplasmic tail. We investigated whether sustained APP signaling during brain development might favor neuronal plasticity and memory process through a direct interaction with the heterotrimeric G-protein subunit GαS (stimulatory G-protein alpha subunit). Our results reveal that APP possesses autonomous regulatory capacity within its intracellular domain that promotes APP cell surface residence, precludes Aβ production, facilitates axodendritic development, and preserves cellular substrates of memory. Altogether, these events contribute to strengthening cognitive functions and are sufficient to modify the course of AD pathology.
Collapse
Affiliation(s)
- Carole Deyts
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Mary Clutter
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Nicholas Pierce
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Thomas B Ladd
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Anna Goddi
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kulandaivelu Vetrivel
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Angèle T Parent
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
7
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
8
|
Guo R, Chen F, Shi Z. Suppression of Notch Signaling Stimulates Progesterone Synthesis by Enhancing the Expression of NR5A2 and NR2F2 in Porcine Granulosa Cells. Genes (Basel) 2020; 11:genes11020120. [PMID: 31978970 PMCID: PMC7073743 DOI: 10.3390/genes11020120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/18/2020] [Indexed: 02/07/2023] Open
Abstract
The conserved Notch pathway is reported to be involved in progesterone synthesis and secretion; however, the exact effects remain controversial. To determine the role and potential mechanisms of the Notch signaling pathway in progesterone biosynthesis in porcine granulosa cells (pGCs), we first used a pharmacological γ-secretase inhibitor, N-(N-(3,5-difluorophenacetyl-l-alanyl))-S-phenylglycine t-butyl ester (DAPT), to block the Notch pathway in cultured pGCs and then evaluated the expression of genes in the progesterone biosynthesis pathway and key transcription factors (TFs) regulating steroidogenesis. We found that DAPT dose- and time-dependently increased progesterone secretion. The expression of steroidogenic proteins NPC1 and StAR and two TFs, NR5A2 and NR2F2, was significantly upregulated, while the expression of HSD3B was significantly downregulated. Furthermore, knockdown of both NR5A2 and NR2F2 with specific siRNAs blocked the upregulatory effects of DAPT on progesterone secretion and reversed the effects of DAPT on the expression of NPC1, StAR, and HSD3B. Moreover, knockdown of NR5A2 and NR2F2 stimulated the expression of Notch3. In conclusion, the inhibition of Notch signaling stimulated progesterone secretion by enhancing the expression of NPC1 and StAR, and the two TFs NR5A2 and NR2F2 acted as downstream TFs of Notch signaling in regulating progesterone synthesis.
Collapse
Affiliation(s)
- Rihong Guo
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Fang Chen
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Zhendan Shi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
- Correspondence:
| |
Collapse
|
9
|
Conrad D, Wilker S, Schneider A, Karabatsiakis A, Pfeiffer A, Kolassa S, Freytag V, Vukojevic V, Vogler C, Milnik A, Papassotiropoulos A, J.‐F. de Quervain D, Elbert T, Kolassa I. Integrated genetic, epigenetic, and gene set enrichment analyses identify NOTCH as a potential mediator for PTSD risk after trauma: Results from two independent African cohorts. Psychophysiology 2020; 57:e13288. [PMID: 30328613 PMCID: PMC7379258 DOI: 10.1111/psyp.13288] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022]
Abstract
The risk of developing posttraumatic stress disorder (PTSD) increases with the number of traumatic event types experienced (trauma load) in interaction with other psychobiological risk factors. The NOTCH (neurogenic locus notch homolog proteins) signaling pathway, consisting of four different trans-membrane receptor proteins (NOTCH1-4), constitutes an evolutionarily well-conserved intercellular communication pathway (involved, e.g., in cell-cell interaction, inflammatory signaling, and learning processes). Its association with fear memory consolidation makes it an interesting candidate for PTSD research. We tested for significant associations of common genetic variants of NOTCH1-4 (investigated by microarray) and genomic methylation of saliva-derived DNA with lifetime PTSD risk in independent cohorts from Northern Uganda (N1 = 924) and Rwanda (N2 = 371), and investigated whether NOTCH-related gene sets were enriched for associations with lifetime PTSD risk. We found associations of lifetime PTSD risk with single nucleotide polymorphism (SNP) rs2074621 (NOTCH3) (puncorrected = 0.04) in both cohorts, and with methylation of CpG site cg17519949 (NOTCH3) (puncorrected = 0.05) in Rwandans. Yet, none of the (epi-)genetic associations survived multiple testing correction. Gene set enrichment analyses revealed enrichment for associations of two NOTCH pathways with lifetime PTSD risk in Ugandans: NOTCH binding (pcorrected = 0.003) and NOTCH receptor processing (pcorrected = 0.01). The environmental factor trauma load was significant in all analyses (all p < 0.001). Our integrated methodological approach suggests NOTCH as a possible mediator of PTSD risk after trauma. The results require replication, and the precise underlying pathophysiological mechanisms should be illuminated in future studies.
Collapse
Affiliation(s)
- Daniela Conrad
- Clinical Psychology and NeuropsychologyUniversity of KonstanzKonstanzGermany
- Clinical & Biological Psychology, Institute of Psychology and EducationUlm UniversityUlmGermany
| | - Sarah Wilker
- Clinical & Biological Psychology, Institute of Psychology and EducationUlm UniversityUlmGermany
| | - Anna Schneider
- Clinical & Biological Psychology, Institute of Psychology and EducationUlm UniversityUlmGermany
| | - Alexander Karabatsiakis
- Clinical & Biological Psychology, Institute of Psychology and EducationUlm UniversityUlmGermany
| | - Anett Pfeiffer
- Clinical Psychology and NeuropsychologyUniversity of KonstanzKonstanzGermany
| | | | - Virginie Freytag
- Division of Molecular NeuroscienceUniversity of BaselBaselSwitzerland
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
| | - Vanja Vukojevic
- Division of Molecular NeuroscienceUniversity of BaselBaselSwitzerland
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
- Department Biozentrum, Life Sciences Training FacilityUniversity of BaselBaselSwitzerland
- Psychiatric University ClinicsUniversity of BaselBaselSwitzerland
| | - Christian Vogler
- Division of Molecular NeuroscienceUniversity of BaselBaselSwitzerland
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
- Psychiatric University ClinicsUniversity of BaselBaselSwitzerland
| | - Annette Milnik
- Division of Molecular NeuroscienceUniversity of BaselBaselSwitzerland
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
- Psychiatric University ClinicsUniversity of BaselBaselSwitzerland
| | - Andreas Papassotiropoulos
- Division of Molecular NeuroscienceUniversity of BaselBaselSwitzerland
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
- Department Biozentrum, Life Sciences Training FacilityUniversity of BaselBaselSwitzerland
- Psychiatric University ClinicsUniversity of BaselBaselSwitzerland
| | - Dominique J.‐F. de Quervain
- Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
- Psychiatric University ClinicsUniversity of BaselBaselSwitzerland
- Division of Cognitive NeuroscienceUniversity of BaselBaselSwitzerland
| | - Thomas Elbert
- Clinical Psychology and NeuropsychologyUniversity of KonstanzKonstanzGermany
| | - Iris‐Tatjana Kolassa
- Clinical & Biological Psychology, Institute of Psychology and EducationUlm UniversityUlmGermany
| |
Collapse
|
10
|
Massimiani M, Lacconi V, La Civita F, Ticconi C, Rago R, Campagnolo L. Molecular Signaling Regulating Endometrium-Blastocyst Crosstalk. Int J Mol Sci 2019; 21:E23. [PMID: 31861484 PMCID: PMC6981505 DOI: 10.3390/ijms21010023] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/29/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Implantation of the embryo into the uterine endometrium is one of the most finely-regulated processes that leads to the establishment of a successful pregnancy. A plethora of factors are released in a time-specific fashion to synchronize the differentiation program of both the embryo and the endometrium. Indeed, blastocyst implantation in the uterus occurs in a limited time frame called the "window of implantation" (WOI), during which the maternal endometrium undergoes dramatic changes, collectively called "decidualization". Decidualization is guided not just by maternal factors (e.g., estrogen, progesterone, thyroid hormone), but also by molecules secreted by the embryo, such as chorionic gonadotropin (CG) and interleukin-1β (IL-1 β), just to cite few. Once reached the uterine cavity, the embryo orients correctly toward the uterine epithelium, interacts with specialized structures, called pinopodes, and begins the process of adhesion and invasion. All these events are guided by factors secreted by both the endometrium and the embryo, such as leukemia inhibitory factor (LIF), integrins and their ligands, adhesion molecules, Notch family members, and metalloproteinases and their inhibitors. The aim of this review is to give an overview of the factors and mechanisms regulating implantation, with a focus on those involved in the complex crosstalk between the blastocyst and the endometrium.
Collapse
Affiliation(s)
- Micol Massimiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro, 8, 00131 Rome, Italy
| | - Valentina Lacconi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| | - Fabio La Civita
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| | - Carlo Ticconi
- Department of Surgical Sciences, Section of Gynecology and Obstetrics, University Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy;
| | - Rocco Rago
- Physiopathology of Reproduction and Andrology Unit, Sandro Pertini Hospital, Via dei Monti Tiburtini 385/389, 00157 Rome, Italy;
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| |
Collapse
|
11
|
Lee YJ, Ch'ng TH. RIP at the Synapse and the Role of Intracellular Domains in Neurons. Neuromolecular Med 2019; 22:1-24. [PMID: 31346933 DOI: 10.1007/s12017-019-08556-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Regulated intramembrane proteolysis (RIP) occurs in a cell when transmembrane proteins are cleaved by intramembrane proteases such as secretases to generate soluble protein fragments in the extracellular environment and the cytosol. In the cytosol, these soluble intracellular domains (ICDs) have local functions near the site of cleavage or in many cases, translocate to the nucleus to modulate gene expression. While the mechanism of RIP is relatively well studied, the fate and function of ICDs for most substrate proteins remain poorly characterized. In neurons, RIP occurs in various subcellular compartments including at the synapse. In this review, we summarize current research on RIP in neurons, focusing specifically on synaptic proteins where the presence and function of the ICDs have been reported. We also briefly discuss activity-driven processing of RIP substrates at the synapse and the cellular machinery that support long-distance transport of ICDs from the synapse to the nucleus. Finally, we describe future challenges in this field of research in the context of understanding the contribution of ICDs in neuronal function.
Collapse
Affiliation(s)
- Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore.,Interdisciplinary Graduate School (IGS), Nanyang Technological University, Singapore, Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
12
|
Landeira BS, Santana TTDS, Araújo JADM, Tabet EI, Tannous BA, Schroeder T, Costa MR. Activity-Independent Effects of CREB on Neuronal Survival and Differentiation during Mouse Cerebral Cortex Development. Cereb Cortex 2019; 28:538-548. [PMID: 27999124 DOI: 10.1093/cercor/bhw387] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/01/2016] [Indexed: 11/14/2022] Open
Abstract
Neuronal survival and morphological maturation depends on the action of the transcription factor calcium responsive element binding protein (CREB), which regulates expression of several target genes in an activity-dependent manner. However, it remains largely unknown whether CREB-mediated transcription could play a role at early stages of neuronal differentiation, prior to the establishment of functional synaptic contacts. Here, we show that CREB is phosphorylated at very early stages of neuronal differentiation in vivo and in vitro, even in the absence of depolarizing agents. Using genetic tools, we also show that inhibition of CREB-signaling affects neuronal growth and survival in vitro without affecting cell proliferation and neurogenesis. Expression of A-CREB or M-CREB, 2 dominant-negative inhibitors of CREB, decreases cell survival and the complexity of neuronal arborization. Similar changes are observed in neurons treated with protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitors, which also show decreased levels of pCREBSer133. Notably, expression of CREB-FY, a Tyr134Phe CREB mutant with a lower Km for phosphorylation, partly rescues the effects of PKA and CaMKII inhibition. Our data indicate that CREB-mediated signaling play important roles at early stages of cortical neuron differentiation, prior to the establishment of fully functional synaptic contacts.
Collapse
Affiliation(s)
| | | | | | - Elie I Tabet
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr. 26, 4058 Basel, Switzerland
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte, Natal 59056-450, Brazil
| |
Collapse
|
13
|
Wang Z, Kawaguchi K, Honda M, Hashimoto S, Shirasaki T, Okada H, Orita N, Shimakami T, Yamashita T, Sakai Y, Mizukoshi E, Murakami S, Kaneko S. Notch signaling facilitates hepatitis B virus covalently closed circular DNA transcription via cAMP response element-binding protein with E3 ubiquitin ligase-modulation. Sci Rep 2019; 9:1621. [PMID: 30733490 PMCID: PMC6367350 DOI: 10.1038/s41598-018-38139-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Notch1 is regulated by E3 ubiquitin ligases, with proteasomal degradation of the Notch intracellular domain affecting the transcription of target genes. cAMP response element-binding protein (CREB) mediates the transcription of hepatitis B virus (HBV) covalently closed circular DNA (cccDNA). We assessed the relationship between HBV cccDNA and Notch signaling activities. HBV cccDNA levels and relative gene expression were evaluated in HBV-replicating cells treated with Jagged1 shRNA and a γ-secretase inhibitor. The effects of these factors in surgically resected clinical samples were also assessed. Notch inhibition suppressed HBV cccDNA and CREB-related expression but increased ITCH and NUMB levels. Proteasome inhibitor augmented HBV cccDNA, restored Notch and CREB expression, and inhibited ITCH and NUMB function. Increased HBV cccDNA was observed after ITCH and NUMB blockage, even after treatment with the adenylate cyclase activator forskolin; protein kinase A (PKA) inhibitor had the opposite effect. Notch activation and E3 ligase inactivation were observed in HBV-positive cells in clinical liver tissue. Collectively, these findings reveal that Notch signaling activity facilitates HBV cccDNA transcription via CREB to trigger the downstream PKA-phospho-CREB cascade and is regulated by E3 ubiquitin ligase-modulation of the Notch intracellular domain.
Collapse
Affiliation(s)
- Zijing Wang
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazunori Kawaguchi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan.
| | - Masao Honda
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Shinichi Hashimoto
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Takayoshi Shirasaki
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hikari Okada
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Noriaki Orita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Tetsuro Shimakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Yoshio Sakai
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Seishi Murakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
14
|
Petruccelli E, Feyder M, Ledru N, Jaques Y, Anderson E, Kaun KR. Alcohol Activates Scabrous-Notch to Influence Associated Memories. Neuron 2018; 100:1209-1223.e4. [PMID: 30482693 DOI: 10.1016/j.neuron.2018.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/17/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Drugs of abuse, like alcohol, modulate gene expression in reward circuits and consequently alter behavior. However, the in vivo cellular mechanisms through which alcohol induces lasting transcriptional changes are unclear. We show that Drosophila Notch/Su(H) signaling and the secreted fibrinogen-related protein Scabrous in mushroom body (MB) memory circuitry are important for the enduring preference of cues associated with alcohol's rewarding properties. Alcohol exposure affects Notch responsivity in the adult MB and alters Su(H) targeting at the dopamine-2-like receptor (Dop2R). Alcohol cue training also caused lasting changes to the MB nuclear transcriptome, including changes in the alternative splicing of Dop2R and newly implicated transcripts like Stat92E. Together, our data suggest that alcohol-induced activation of the highly conserved Notch pathway and accompanying transcriptional responses in memory circuitry contribute to addiction. Ultimately, this provides mechanistic insight into the etiology and pathophysiology of alcohol use disorder.
Collapse
Affiliation(s)
- Emily Petruccelli
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Michael Feyder
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Nicolas Ledru
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Yanabah Jaques
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Edward Anderson
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
15
|
Chiappara G, Sciarrino S, Di Sano C, Gallina S, Speciale R, Lorusso F, Di Vincenzo S, D'Anna C, Bruno A, Gjomarkaj M, Pace E. Notch-1 signaling activation sustains overexpression of interleukin 33 in the epithelium of nasal polyps. J Cell Physiol 2018; 234:4582-4596. [PMID: 30259982 DOI: 10.1002/jcp.27237] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Alterations in the nasal epithelial barrier homeostasis and increased interleukin 33 (IL-33) expression contribute to the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP). AIMS As Notch-1 signaling is crucial in repair processes of mucosa, the current study assessed Notch-1/Jagged-1 signaling and IL-33 in the epithelium of nasal polyps biopsies from allergic (A-CRSwNP; n = 9) and not allergic (NA-CRSwNP; n = 9) subjects by immunohistochemistry. We also assessed, in a model of nasal epithelial cells, the effects of stimulation of Notch-1 with Jagged-1 on the expression of IL-33 (by flow cytometry, immunofluorescence, and immunocytochemistry), Jagged-1 (by flow cytometry), and p-CREB transcription factor (by western blot analysis). RESULTS Ex vivo (a) in normal epithelium, the expression of Notch-1 and IL-33 were higher in NA-CRSwNP than in A-CRSwNP; (b) in metaplastic epithelium, the expression of Notch-1, Jagged-1, and IL-33 were higher in NA-CRSwNP than in A-CRSwNP; (c) in hyperplastic epithelium, the expression of Notch-1, Jagged-1, and IL-33 were higher in A-CRSwNP than in NA-CRSwNP; and (d) in basal epithelial cells, no differences were observed in the expression of Jagged-1, IL-33, and Notch-1. The expression of Notch-1 significantly correlated with the expression of IL-33. In vitro, stimulation of Notch-1 with Jagged-1 induced the expression of (a) Jagged-1; (b) IL-33; and (c) p-CREB transcription factor. The inhibitor of Notch-1, DAPT, reduced all the effects of Jagged-1 on nasal epithelial cells. CONCLUSIONS The data herein provided support, for the first time, a putative role of Notch-1/Jagged-1 signaling in the overexpression of IL-33 in the epithelium of nasal polyps from patients with CRSwNP.
Collapse
Affiliation(s)
- G Chiappara
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - S Sciarrino
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - C Di Sano
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - S Gallina
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, Sezione di Otorinolaringoiatria, Università degli Studi di Palermo, Palermo, Italy
| | - R Speciale
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, Sezione di Otorinolaringoiatria, Università degli Studi di Palermo, Palermo, Italy
| | - F Lorusso
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, Sezione di Otorinolaringoiatria, Università degli Studi di Palermo, Palermo, Italy
| | - S Di Vincenzo
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - C D'Anna
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - A Bruno
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - M Gjomarkaj
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - E Pace
- Istituto di Biomedicina e Immunologia Molecolare, Dipartimento di Biomedicina, Consiglio Nazionale delle Ricerche, Palermo, Italy
| |
Collapse
|
16
|
A role for APP in Wnt signalling links synapse loss with β-amyloid production. Transl Psychiatry 2018; 8:179. [PMID: 30232325 PMCID: PMC6145937 DOI: 10.1038/s41398-018-0231-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 01/18/2023] Open
Abstract
In Alzheimer's disease (AD), the canonical Wnt inhibitor Dickkopf-1 (Dkk1) is induced by β-amyloid (Aβ) and shifts the balance from canonical towards non-canonical Wnt signalling. Canonical (Wnt-β-catenin) signalling promotes synapse stability, while non-canonical (Wnt-PCP) signalling favours synapse retraction; thus Aβ-driven synapse loss is mediated by Dkk1. Here we show that the Amyloid Precursor Protein (APP) co-activates both arms of Wnt signalling through physical interactions with Wnt co-receptors LRP6 and Vangl2, to bi-directionally modulate synapse stability. Furthermore, activation of non-canonical Wnt signalling enhances Aβ production, while activation of canonical signalling suppresses Aβ production. Together, these findings identify a pathogenic-positive feedback loop in which Aβ induces Dkk1 expression, thereby activating non-canonical Wnt signalling to promote synapse loss and drive further Aβ production. The Swedish familial AD variant of APP (APPSwe) more readily co-activates non-canonical, at the expense of canonical Wnt activity, indicating that its pathogenicity likely involves direct effects on synapses, in addition to increased Aβ production. Finally, we report that pharmacological inhibition of the Aβ-Dkk1-Aβ positive feedback loop with the drug fasudil can restore the balance between Wnt pathways, prevent dendritic spine withdrawal in vitro, and reduce Aβ load in vivo in mice with advanced amyloid pathology. These results clarify a relationship between Aβ accumulation and synapse loss and provide direction for the development of potential disease-modifying treatments.
Collapse
|
17
|
Garcia-Concejo A, Jimenez-Gonzalez A, Rodriguez RE. Opioid and Notch signaling pathways are reciprocally regulated through miR- 29a and miR-212 expression. Biochim Biophys Acta Gen Subj 2018; 1862:2605-2612. [PMID: 30251655 DOI: 10.1016/j.bbagen.2018.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND The abuse of opioids, such as morphine and phentanyl or other drugs as heroin is a social and health problem that affects an increasing number of people each year. The activation of the mu opioid receptor triggers several molecular changes that alter the expression of diverse genes, including miRNAs. The dysregulation of these molecules could explain some of the developmental alterations that are induced after drug intake. In addition, the Notch signaling cascade has also been related to alterations on these processes. METHODS Zebrafish embryos and SH-SY5Y cells were used to assess the effects of opioid and Notch signaling on the expression on miR-29a and miR-212/132 by qPCR and ChIP-qPCR. Notch1 expression was analyzed using in situ hybridization on 24 hpf zebrafish embryos. In addition, OPRM1 and NICD levels were measured using western blot on the cultured cells to determine the cross-talk between the two pathways. RESULTS We have observed changes in the levels of miR-212/132 after administrating DAPT to zebrafish embryos indicating that this pathway could be regulating mu opioid receptor expression. In addition, the ISH experiment showed changes in Notch1 expression after morphine and DAPT administration. Moreover, morphine affects the expression of miR-29a through NF-κB, therefore controlling the cleavage and activation of Notch through ADAM12 expression. CONCLUSIONS This study shows that these two pathways are closely related, and could explain the alterations triggered in the early stages of the development of addiction. GENERAL SIGNIFICANCE Opioid and Notch pathway are reciprocally regulated by the miRNAs 212/132 and 29a.
Collapse
Affiliation(s)
- Adrian Garcia-Concejo
- Institute of Neurosciences of Castilla y Leon (INCyL), C/Pintor Fernando Gallego, 1, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Edificio Virgen de la Vega. Décima Planta, P° de San Vicente 58-182, 37007 Salamanca, Spain
| | - Ada Jimenez-Gonzalez
- Institute of Neurosciences of Castilla y Leon (INCyL), C/Pintor Fernando Gallego, 1, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Edificio Virgen de la Vega. Décima Planta, P° de San Vicente 58-182, 37007 Salamanca, Spain
| | - Raquel E Rodriguez
- Institute of Neurosciences of Castilla y Leon (INCyL), C/Pintor Fernando Gallego, 1, 37007 Salamanca, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, C/Alfonso X El Sabio, 0 S-N Campus Miguel De Unamuno, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Edificio Virgen de la Vega. Décima Planta, P° de San Vicente 58-182, 37007 Salamanca, Spain.
| |
Collapse
|
18
|
Chronic Hippocampal Expression of Notch Intracellular Domain Induces Vascular Thickening, Reduces Glucose Availability, and Exacerbates Spatial Memory Deficits in a Rat Model of Early Alzheimer. Mol Neurobiol 2018; 55:8637-8650. [PMID: 29582397 DOI: 10.1007/s12035-018-1002-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/07/2018] [Indexed: 02/07/2023]
Abstract
The specific roles of Notch in progressive adulthood neurodegenerative disorders have begun to be unraveled in recent years. A number of independent studies have shown significant increases of Notch expression in brains from patients at later stages of sporadic Alzheimer's disease (AD). However, the impact of Notch canonical signaling activation in the pathophysiology of AD is still elusive. To further investigate this issue, 2-month-old wild-type (WT) and hemizygous McGill-R-Thy1-APP rats (Tg(+/-)) were injected in CA1 with lentiviral particles (LVP) expressing the transcriptionally active fragment of Notch, known as Notch Intracellular Domain (NICD), (LVP-NICD), or control lentivirus particles (LVP-C). The Tg(+/-) rat model captures presymptomatic aspects of the AD pathology, including intraneuronal amyloid beta (Aβ) accumulation and early cognitive deficits. Seven months after LVP administration, Morris water maze test was performed, and brains isolated for biochemical and histological analysis. Our results showed a learning impairment and a worsening of spatial memory in LVP-NICD- as compared to LVP-C-injected Tg(+/-) rats. In addition, immuno histochemistry, ELISA multiplex, Western blot, RT-qPCR, and 1H-NMR spectrometry of cerebrospinal fluid (CSF) indicated that chronic expression of NICD promoted hippocampal vessel thickening with accumulation of Aβ in brain microvasculature, alteration of blood-brain barrier (BBB) permeability, and a decrease of CSF glucose levels. These findings suggest that, in the presence of early Aβ pathology, expression of NICD may contribute to the development of microvascular abnormalities, altering glucose transport at the BBB with impact on early decline of spatial learning and memory.
Collapse
|
19
|
Chiola S, Do MD, Centrone L, Mallamaci A. Foxg1 Overexpression in Neocortical Pyramids Stimulates Dendrite Elongation Via Hes1 and pCreb1 Upregulation. Cereb Cortex 2018; 29:1006-1019. [DOI: 10.1093/cercor/bhy007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/07/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Simone Chiola
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Mihn Duc Do
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Lucy Centrone
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Antonello Mallamaci
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| |
Collapse
|
20
|
Activity-dependent synapse to nucleus signaling. Neurobiol Learn Mem 2017; 138:78-84. [DOI: 10.1016/j.nlm.2016.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/20/2016] [Accepted: 07/23/2016] [Indexed: 11/15/2022]
|
21
|
Hutter G, Sailer M, Azad TD, von Bueren AO, Nollau P, Frank S, Tostado C, Sarvepalli D, Ghosh A, Ritz MF, Boulay JL, Mariani L. Reverse phase protein arrays enable glioblastoma molecular subtyping. J Neurooncol 2016; 131:437-448. [PMID: 27858266 DOI: 10.1007/s11060-016-2316-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/06/2016] [Indexed: 12/24/2022]
Abstract
In the present study we investigated the phosphorylation status of the 12 most important signaling cascades in glioblastomas. More than 60 tumor and control biopsies from tumor center and periphery (based on neuronavigation) were subjected to selective protein expression analysis using reverse-phase protein arrays (RPPA) incubated with antibodies against posttranslationally modified cancer pathway proteins. The ratio between phosphorylated (or modified) and non-phosphorylated protein was assessed. All samples were histopathologically validated and proteomic profiles correlated with clinical and survival data. By RPPA, we identified three distinct activation patterns within glioblastoma defined by the ratios of pCREB1/CREB1, NOTCH-ICD/NOTCH1, and pGSK3β/GSK3β, respectively. These subclasses demonstrated distinct overall survival patterns in a cohort of patients from a single-institution and in an analysis of publicly available data. In particular, a high pGSK3β/GSK3β-ratio was associated with a poor survival. Wnt-activation/GSK3β-inhibition in U373 and U251 cell lines halted glioma cell proliferation and migration. Gene expression analysis was used as an internal quality control of baseline proteomic data. The protein expression and phosphorylation had a higher resolution, resulting in a better class-subdivision than mRNA based stratification data. Patients with different proteomic profiles from multiple biopsies showed a worse overall survival. The CREB1-, NOTCH1-, GSK3β-phosphorylation status correlated with glioma grades. RPPA represent a fast and reliable tool to supplement morphological diagnosis with pathway-specific information in individual tumors. These data can be exploited for molecular stratification and possible combinatorial treatment planning. Further, our results may optimize current glioma grading algorithms.
Collapse
Affiliation(s)
- Gregor Hutter
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland. .,Department of Neurosurgery, Stanford University, 300 Pasteur Drive, Stanford, 94305, CA, USA.
| | - Martin Sailer
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Tej Deepak Azad
- Department of Neurosurgery, Stanford University, 300 Pasteur Drive, Stanford, 94305, CA, USA
| | - André O von Bueren
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Hospital of Geneva, Geneva, Switzerland.,CANSEARCH Research Laboratory, Department of Pediatrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Nollau
- Diagnostic Center, Institute for Clinical Chemistry, University Medical Center Hamburg-Eppendorf (UKE), CAMPUS Research Building N27, Martinistraße 52, 20246, Hamburg, Germany
| | - Stephan Frank
- Division of Neuro- and Ophthalmopathology, Department of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Cristobal Tostado
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Durga Sarvepalli
- Molecular Signalling and Gene Therapy, Narayana Nethralaya, Narayana Health City, # 258/A, Bommasandra, Hosur Road, Bangalore, 560 099, India
| | - Arkasubhra Ghosh
- Molecular Signalling and Gene Therapy, Narayana Nethralaya, Narayana Health City, # 258/A, Bommasandra, Hosur Road, Bangalore, 560 099, India
| | - Marie-Françoise Ritz
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Jean-Louis Boulay
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Luigi Mariani
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| |
Collapse
|
22
|
Golovin RM, Broadie K. Developmental experience-dependent plasticity in the first synapse of the Drosophila olfactory circuit. J Neurophysiol 2016; 116:2730-2738. [PMID: 27683892 DOI: 10.1152/jn.00616.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/26/2016] [Indexed: 12/15/2022] Open
Abstract
Evidence accumulating over the past 15 years soundly refutes the dogma that the Drosophila nervous system is hardwired. The preponderance of studies reveals activity-dependent neural circuit refinement driving optimization of behavioral outputs. We describe developmental, sensory input-dependent plasticity in the brain olfactory antennal lobe, which we term long-term central adaption (LTCA). LTCA is evoked by prolonged exposure to an odorant during the first week of posteclosion life, resulting in a persistently decreased response to aversive odors and an enhanced response to attractive odors. This limited window of early-use, experience-dependent plasticity represents a critical period of olfactory circuit refinement tuned by initial sensory input. Consequent behavioral adaptations have been associated with changes in the output of olfactory projection neurons to higher brain centers. Recent studies have indicated a central role for local interneuron signaling in LTCA presentation. Genetic and molecular analyses have implicated the mRNA-binding fragile X mental retardation protein and ataxin-2 regulators, Notch trans-synaptic signaling, and cAMP signal transduction as core regulatory steps driving LTCA. In this article, we discuss the structural, functional, and behavioral changes associated with LTCA and review our current understanding of the molecular pathways underlying these developmental, experience-dependent changes in the olfactory circuitry.
Collapse
Affiliation(s)
- Randall M Golovin
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; and
| | - Kendal Broadie
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; and .,Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
23
|
Aberrant activation of canonical Notch1 signaling in the mouse uterus decreases progesterone receptor by hypermethylation and leads to infertility. Proc Natl Acad Sci U S A 2016; 113:2300-5. [PMID: 26858409 DOI: 10.1073/pnas.1520441113] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In mammalian reproduction, implantation is one of the most critical events. Failure of implantation and the subsequent decidualization contribute to more than 75% of pregnancy losses in women. Our laboratory has previously reported that inhibition of Notch signaling results in impaired decidualization in both women and a transgenic mouse model. In this study, we generated a Notch gain-of-function transgenic mouse by conditionally overexpressing the Notch1 intracellular domain (N1ICD) in the reproductive tract driven by a progesterone receptor (Pgr) -Cre. We show that the overexpression of N1ICD in the uterus results in complete infertility as a consequence of multiple developmental and physiological defects, including the absence of uterine glands and dysregulation of progesterone and estrogen signaling by a Recombination Signal Binding Protein Jκ-dependent signaling mechanism. We further show that the inhibition of progesterone signaling is caused by hypermethylation of its receptor Pgr by Notch1 overexpression through the transcription factor PU.1 and DNA methyltransferase 3b (Dnmt3b). We have generated a mouse model to study the consequence of increased Notch signaling in female reproduction and provide the first evidence, to our knowledge, that Notch signaling can regulate epigenetic modification of the Pgr.
Collapse
|
24
|
Zhang J, Yin JCP, Wesley CS. Notch Intracellular Domain (NICD) Suppresses Long-Term Memory Formation in Adult Drosophila Flies. Cell Mol Neurobiol 2015; 35:763-8. [PMID: 25791355 DOI: 10.1007/s10571-015-0183-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/16/2015] [Indexed: 01/09/2023]
Abstract
Notch receptor signaling is evolutionarily conserved and well known for its roles in animal development. Many studies in Drosophila have shown that Notch also performs important functions in memory formation in adult flies. An intriguing observation is that increased expression of the full-length Notch receptor (Nfull) triggers long-term memory (LTM) formation even after very weak training (single training). Canonical Notch signaling is mediated by Notch intracellular domain (NICD), but it is not known whether increased expression of NICD recapitulates the LTM enhancement induced by increased Nfull expression. Here, we report that increased NICD expression either has no impact on LTM formation or suppresses it. Furthermore, it either has no impact or decreases both the levels and activity of cAMP response element binding protein, a key factor supporting LTM. These results indicate that NICD signaling is not sufficient to explain Nfull-induced LTM enhancement. Our findings may also shed light on the molecular mechanisms of memory loss in neurological diseases associated with increased NICD expression and canonical Notch signaling.
Collapse
Affiliation(s)
- Jiabin Zhang
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | | |
Collapse
|
25
|
Su RW, Fazleabas AT. Implantation and Establishment of Pregnancy in Human and Nonhuman Primates. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2015; 216:189-213. [PMID: 26450500 PMCID: PMC5098399 DOI: 10.1007/978-3-319-15856-3_10] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Implantation and the establishment of pregnancy are critical for the propagation of the species, but yet remain the limiting steps in human and primate reproduction. Successful implantation requires a competent blastocyst and a receptive endometrium during a specific window of time during the menstrual cycle to initiate the bilateral communication required for the establishment of a successful pregnancy. This chapter provides an overview of these processes and discusses the molecular mechanisms associated with implantation of the blastocyst and decidualization of the uterus in primates.
Collapse
Affiliation(s)
- Ren-Wei Su
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA.
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| |
Collapse
|