1
|
Zhang D, Tu H, Hu W, Li Y, Wadman MC, Li YL. Hydrogen Peroxide-Induced Re-Expression of Repressor Element 1-Silencing Transcription Factor Contributes to Cardiac Vagal Dysfunction in Type 2 Diabetes Mellitus. Antioxidants (Basel) 2025; 14:588. [PMID: 40427470 DOI: 10.3390/antiox14050588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Diabetes mellitus, especially type 2 diabetes mellitus (T2DM), is a major health problem worldwide and has become a leading cause of mortality. As a common complication of patients with T2DM, cardiac autonomic dysfunction (including sympathetic overactivation and reduced vagal tone) is associated with a higher risk of arrhythmia-related sudden cardiac death. Our previous study found that T2DM-elevated hydrogen peroxide (H2O2) levels in atrioventricular ganglion (AVG) neurons contribute to the decrease in cardiac vagal function and ventricular arrhythmogenesis through inhibition of N-type Ca2+ channels (Cav2.2). In the present study, treatment with exogenous H2O2 in differentiated NG108-15 cells increased REST expression and decreased Cav2.2-α expression. Adenoviral catalase gene transfection into the AVG neurons significantly reduced the REST levels elevated by a high-fat diet plus streptozotocin-induced T2DM. Lentiviral REST shRNA transfection markedly increased Cav2.2-α expression in the AVG neurons from T2DM rats. REST shRNA also activated N-type Ca2+ channels and increased cell excitability of AVG neurons in T2DM rats. Additionally, REST shRNA markedly improved cardiac vagal activation in T2DM rats. The present study suggests that the H2O2-REST-Cav2.2 channel signaling axis could be a potential therapeutic target to normalize cardiac vagal dysfunction and its related cardiac complications in T2DM.
Collapse
Affiliation(s)
- Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wenfeng Hu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yu Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael C Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Kulkarni GC, Saha R, Peters CJ. Ion channel expression and function in glioblastoma multiforme (GBM): pathophysiological mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119982. [PMID: 40328081 DOI: 10.1016/j.bbamcr.2025.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
Glioblastoma Multiforme (GBM) is a highly malignant and diffusely invasive WHO Grade IV brain tumor arising from glial and neural stem cells. GBM is characterized by rapid proliferation and migration, aggressive invasion of local brain parenchyma, a hypoxic microenvironment, resistance to apoptosis and high vascular remodeling and angiogenesis. These hallmarks contribute to a near universal tumor recurrence after treatment or resection and poor patient prognosis. Ion channels, a superfamily of proteins responsible for permitting ion flux across otherwise impermeant membranes, show extensive remodeling in GBM with aberrant function mechanistically linked to manipulation of each of these hallmarks. In this review, we will discuss the known links between ion channel expression and activity and cellular processes that are enhanced or perturbed during GBM formation or progression. We will also discuss the extent to which basic or translational findings on ion channels in GBM samples or cell lines have shown preclinical promise towards the development of improved therapeutics against GBMs.
Collapse
Affiliation(s)
- Gauri C Kulkarni
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Rayna Saha
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Christian J Peters
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Szymanowicz O, Drużdż A, Słowikowski B, Pawlak S, Potocka E, Goutor U, Konieczny M, Ciastoń M, Lewandowska A, Jagodziński PP, Kozubski W, Dorszewska J. A Review of the CACNA Gene Family: Its Role in Neurological Disorders. Diseases 2024; 12:90. [PMID: 38785745 PMCID: PMC11119137 DOI: 10.3390/diseases12050090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Calcium channels are specialized ion channels exhibiting selective permeability to calcium ions. Calcium channels, comprising voltage-dependent and ligand-gated types, are pivotal in neuronal function, with their dysregulation is implicated in various neurological disorders. This review delves into the significance of the CACNA genes, including CACNA1A, CACNA1B, CACNA1C, CACNA1D, CACNA1E, CACNA1G, and CACNA1H, in the pathogenesis of conditions such as migraine, epilepsy, cerebellar ataxia, dystonia, and cerebellar atrophy. Specifically, variants in CACNA1A have been linked to familial hemiplegic migraine and epileptic seizures, underscoring its importance in neurological disease etiology. Furthermore, different genetic variants of CACNA1B have been associated with migraine susceptibility, further highlighting the role of CACNA genes in migraine pathology. The complex relationship between CACNA gene variants and neurological phenotypes, including focal seizures and ataxia, presents a variety of clinical manifestations of impaired calcium channel function. The aim of this article was to explore the role of CACNA genes in various neurological disorders, elucidating their significance in conditions such as migraine, epilepsy, and cerebellar ataxias. Further exploration of CACNA gene variants and their interactions with molecular factors, such as microRNAs, holds promise for advancing our understanding of genetic neurological disorders.
Collapse
Affiliation(s)
- Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Artur Drużdż
- Department of Neurology, Municipal Hospital in Poznan, 61-285 Poznan, Poland;
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Sandra Pawlak
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Ewelina Potocka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Ulyana Goutor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Mateusz Konieczny
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Małgorzata Ciastoń
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Aleksandra Lewandowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| |
Collapse
|
4
|
Najafi P, Reimer C, Gilthorpe JD, Jacobsen KR, Ramløse M, Paul NF, Simianer H, Tetens J, Falker-Gieske C. Genomic evidence for the suitability of Göttingen Minipigs with a rare seizure phenotype as a model for human epilepsy. Neurogenetics 2024; 25:103-117. [PMID: 38383918 PMCID: PMC11076379 DOI: 10.1007/s10048-024-00750-2] [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: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024]
Abstract
Epilepsy is a complex genetic disorder that affects about 2% of the global population. Although the frequency and severity of epileptic seizures can be reduced by a range of pharmacological interventions, there are no disease-modifying treatments for epilepsy. The development of new and more effective drugs is hindered by a lack of suitable animal models. Available rodent models may not recapitulate all key aspects of the disease. Spontaneous epileptic convulsions were observed in few Göttingen Minipigs (GMPs), which may provide a valuable alternative animal model for the characterisation of epilepsy-type diseases and for testing new treatments. We have characterised affected GMPs at the genome level and have taken advantage of primary fibroblast cultures to validate the functional impact of fixed genetic variants on the transcriptome level. We found numerous genes connected to calcium metabolism that have not been associated with epilepsy before, such as ADORA2B, CAMK1D, ITPKB, MCOLN2, MYLK, NFATC3, PDGFD, and PHKB. Our results have identified two transcription factor genes, EGR3 and HOXB6, as potential key regulators of CACNA1H, which was previously linked to epilepsy-type disorders in humans. Our findings provide the first set of conclusive results to support the use of affected subsets of GMPs as an alternative and more reliable model system to study human epilepsy. Further neurological and pharmacological validation of the suitability of GMPs as an epilepsy model is therefore warranted.
Collapse
Affiliation(s)
- Pardis Najafi
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Christian Reimer
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Höltystr. 10, 31535, Neustadt, Germany
| | - Jonathan D Gilthorpe
- Department of Integrative Medical Biology, Umeå University, 901 87, Umeå, Sweden
| | - Kirsten R Jacobsen
- Ellegaard Göttingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Maja Ramløse
- Ellegaard Göttingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Nora-Fabienne Paul
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
| | - Henner Simianer
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Jens Tetens
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany.
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany.
| |
Collapse
|
5
|
Leandrou E, Chalatsa I, Anagnostou D, Machalia C, Semitekolou M, Filippa V, Makridakis M, Vlahou A, Anastasiadou E, Vekrellis K, Emmanouilidou E. α-Synuclein oligomers potentiate neuroinflammatory NF-κB activity and induce Ca v3.2 calcium signaling in astrocytes. Transl Neurodegener 2024; 13:11. [PMID: 38378800 PMCID: PMC10880263 DOI: 10.1186/s40035-024-00401-4] [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: 08/28/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND It is now realized that Parkinson's disease (PD) pathology extends beyond the substantia nigra, affecting both central and peripheral nervous systems, and exhibits a variety of non-motor symptoms often preceding motor features. Neuroinflammation induced by activated microglia and astrocytes is thought to underlie these manifestations. α-Synuclein aggregation has been linked with sustained neuroinflammation in PD, aggravating neuronal degeneration; however, there is still a lack of critical information about the structural identity of the α-synuclein conformers that activate microglia and/or astrocytes and the molecular pathways involved. METHODS To investigate the role of α-synuclein conformers in the development and maintenance of neuroinflammation, we used primary quiescent microglia and astrocytes, post-mortem brain tissues from PD patients and A53T α-synuclein transgenic mice that recapitulate key features of PD-related inflammatory responses in the absence of cell death, i.e., increased levels of pro-inflammatory cytokines and complement proteins. Biochemical and -omics techniques including RNAseq and secretomic analyses, combined with 3D reconstruction of individual astrocytes and live calcium imaging, were used to uncover the molecular mechanisms underlying glial responses in the presence of α-synuclein oligomers in vivo and in vitro. RESULTS We found that the presence of SDS-resistant hyper-phosphorylated α-synuclein oligomers, but not monomers, was correlated with sustained inflammatory responses, such as elevated levels of endogenous antibodies and cytokines and microglial activation. Similar oligomeric α-synuclein species were found in post-mortem human brain samples of PD patients but not control individuals. Detailed analysis revealed a decrease in Iba1Low/CD68Low microglia and robust alterations in astrocyte number and morphology including process retraction. Our data indicated an activation of the p38/ATF2 signaling pathway mostly in microglia and a sustained induction of the NF-κB pathway in astrocytes of A53T mice. The sustained NF-κB activity triggered the upregulation of astrocytic T-type Cav3.2 Ca2+ channels, altering the astrocytic secretome and promoting the secretion of IGFBPL1, an IGF-1 binding protein with anti-inflammatory and neuroprotective potential. CONCLUSIONS Our work supports a causative link between the neuron-produced α-synuclein oligomers and sustained neuroinflammation in vivo and maps the signaling pathways that are stimulated in microglia and astrocytes. It also highlights the recruitment of astrocytic Cav3.2 channels as a potential neuroprotective mediator against the α-synuclein-induced neuroinflammation.
Collapse
Affiliation(s)
- Emmanouela Leandrou
- Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15772, Athens, Greece
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Ioanna Chalatsa
- Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15772, Athens, Greece
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Dimitrios Anagnostou
- Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15772, Athens, Greece
| | - Christina Machalia
- Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15772, Athens, Greece
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Maria Semitekolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
- School of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Vicky Filippa
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Manousos Makridakis
- Center for Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Antonia Vlahou
- Center for Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Ema Anastasiadou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Kostas Vekrellis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Evangelia Emmanouilidou
- Department of Chemistry, School of Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15772, Athens, Greece.
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece.
| |
Collapse
|
6
|
Galvis-Montes DS, van Loo KMJ, van Waardenberg AJ, Surges R, Schoch S, Becker AJ, Pitsch J. Highly dynamic inflammatory and excitability transcriptional profiles in hippocampal CA1 following status epilepticus. Sci Rep 2023; 13:22187. [PMID: 38092829 PMCID: PMC10719343 DOI: 10.1038/s41598-023-49310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
Transient brain insults including status epilepticus (SE) can initiate a process termed 'epileptogenesis' that results in chronic temporal lobe epilepsy. As a consequence, the entire tri-synaptic circuit of the hippocampus is fundamentally impaired. A key role in epileptogenesis has been attributed to the CA1 region as the last relay station in the hippocampal circuit and as site of aberrant plasticity, e.g. mediated by acquired channelopathies. The transcriptional profiles of the distinct hippocampal neurons are highly dynamic during epileptogenesis. Here, we aimed to elucidate the early SE-elicited mRNA signature changes and the respective upstream regulatory cascades in CA1. RNA sequencing of CA1 was performed in the mouse pilocarpine-induced SE model at multiple time points ranging from 6 to 72 h after the initial insult. Bioinformatics was used to decipher altered gene expression, signalling cascades and their corresponding cell type profiles. Robust transcriptomic changes were detected at 6 h after SE and at subsequent time points during early epileptogenesis. Major differentially expressed mRNAs encoded primarily immediate early and excitability-related gene products, as well as genes encoding immune signalling factors. Binding sites for the transcription factors Nfkb1, Spi1, Irf8, and two Runx family members, were enriched within promoters of differentially expressed genes related to major inflammatory processes, whereas the transcriptional repressors Suz12, Nfe2l2 and Rest were associated with hyperexcitability and GABA / glutamate receptor activity. CA1 quickly responds to SE by inducing transcription of genes linked to inflammation and excitation stress. Transcription factors mediating this transcriptomic switch represent targets for new highly selected, cell type and time window-specific anti-epileptogenic strategies.
Collapse
Grants
- SCHO 820/4-1, SCHO 820/6-1, SCHO 820/7-1, SCHO 820/5-2, SPP1757, SFB1089, FOR 2715 Deutsche Forschungsgemeinschaft
- SCHO 820/4-1, SCHO 820/6-1, SCHO 820/7-1, SCHO 820/5-2, SPP1757, SFB1089, FOR 2715 Deutsche Forschungsgemeinschaft
- Promotionskolleg 'NeuroImmunology' Else Kröner-Fresenius-Stiftung
- Promotionskolleg 'NeuroImmunology' Else Kröner-Fresenius-Stiftung
- BONFOR program of the Medical Faculty, University of Bonn
- Rheinische Friedrich-Wilhelms-Universität Bonn (1040)
Collapse
Affiliation(s)
- Daniel S Galvis-Montes
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Karen M J van Loo
- Department of Epileptology, Neurology, RWTH Aachen University, Aachen, Germany
| | | | - Rainer Surges
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Susanne Schoch
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Julika Pitsch
- Department of Epileptology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
7
|
Zhang H, Li H, Lu M, Wang S, Ma X, Wang F, Liu J, Li X, Yang H, Zhang F, Shen H, Buckley NJ, Gamper N, Yamoah EN, Lv P. Repressor element 1-silencing transcription factor deficiency yields profound hearing loss through K v7.4 channel upsurge in auditory neurons and hair cells. eLife 2022; 11:76754. [PMID: 36125121 PMCID: PMC9525063 DOI: 10.7554/elife.76754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
Repressor element 1-silencing transcription factor (REST) is a transcriptional repressor that recognizes neuron-restrictive silencer elements in the mammalian genomes in a tissue- and cell-specific manner. The identity of REST target genes and molecular details of how REST regulates them are emerging. We performed conditional null deletion of Rest (cKO), mainly restricted to murine hair cells (HCs) and auditory neurons (aka spiral ganglion neurons [SGNs]). Null inactivation of full-length REST did not affect the development of normal HCs and SGNs but manifested as progressive hearing loss in adult mice. We found that the inactivation of REST resulted in an increased abundance of Kv7.4 channels at the transcript, protein, and functional levels. Specifically, we found that SGNs and HCs from Rest cKO mice displayed increased Kv7.4 expression and augmented Kv7 currents; SGN’s excitability was also significantly reduced. Administration of a compound with Kv7.4 channel activator activity, fasudil, recapitulated progressive hearing loss in mice. In contrast, inhibition of the Kv7 channels by XE991 rescued the auditory phenotype of Rest cKO mice. Previous studies identified some loss-of-function mutations within the Kv7.4-coding gene, Kcnq4, as a causative factor for progressive hearing loss in mice and humans. Thus, the findings reveal that a critical homeostatic Kv7.4 channel level is required for proper auditory functions.
Collapse
Affiliation(s)
- Haiwei Zhang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Hongchen Li
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Mingshun Lu
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Shengnan Wang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Xueya Ma
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Fei Wang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Jiaxi Liu
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Xinyu Li
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Haichao Yang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Fan Zhang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Hebei, China
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Nikita Gamper
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada Reno, Reno, United States
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Hebei, China
| |
Collapse
|
8
|
Huzard D, Martin M, Maingret F, Chemin J, Jeanneteau F, Mery PF, Fossat P, Bourinet E, François A. The impact of C-tactile low-threshold mechanoreceptors on affective touch and social interactions in mice. SCIENCE ADVANCES 2022; 8:eabo7566. [PMID: 35767616 PMCID: PMC9242590 DOI: 10.1126/sciadv.abo7566] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Affective touch is necessary for proper neurodevelopment and sociability. However, it remains unclear how the neurons innervating the skin detect affective and social behaviors. The C low-threshold mechanoreceptors (C-LTMRs), a specific population of somatosensory neurons in mice, appear particularly well suited, physiologically and anatomically, to perceive affective and social touch. However, their contribution to sociability has not been resolved yet. Our observations revealed that C-LTMR functional deficiency induced social isolation and reduced tactile interactions in adulthood. Conversely, transient increase in C-LTMR excitability in adults, using chemogenetics, was rewarding, promoted touch-seeking behaviors, and had prosocial influences on group dynamics. This work provides the first empirical evidence that specific peripheral inputs alone can drive complex social behaviors. It demonstrates the existence of a specialized neuronal circuit, originating in the skin, wired to promote interactions with other individuals.
Collapse
Affiliation(s)
- Damien Huzard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Miquel Martin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - François Maingret
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Jean Chemin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierre-François Mery
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pascal Fossat
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Amaury François
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Corresponding author.
| |
Collapse
|
9
|
SCN1A overexpression, associated with a genomic region marked by a risk variant for a common epilepsy, raises seizure susceptibility. Acta Neuropathol 2022; 144:107-127. [PMID: 35551471 PMCID: PMC9217876 DOI: 10.1007/s00401-022-02429-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/01/2022]
Abstract
Mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures is associated with common variation at rs7587026, located in the promoter region of SCN1A. We sought to explore possible underlying mechanisms. SCN1A expression was analysed in hippocampal biopsy specimens of individuals with mesial temporal lobe epilepsy with hippocampal sclerosis who underwent surgical treatment, and hippocampal neuronal cell loss was quantitatively assessed using immunohistochemistry. In healthy individuals, hippocampal volume was measured using MRI. Analyses were performed stratified by rs7587026 type. To study the functional consequences of increased SCN1A expression, we generated, using transposon-mediated bacterial artificial chromosome transgenesis, a zebrafish line expressing exogenous scn1a, and performed EEG analysis on larval optic tecta at 4 day post-fertilization. Finally, we used an in vitro promoter analysis to study whether the genetic motif containing rs7587026 influences promoter activity. Hippocampal SCN1A expression differed by rs7587026 genotype (Kruskal-Wallis test P = 0.004). Individuals homozygous for the minor allele showed significantly increased expression compared to those homozygous for the major allele (Dunn's test P = 0.003), and to heterozygotes (Dunn's test P = 0.035). No statistically significant differences in hippocampal neuronal cell loss were observed between the three genotypes. Among 597 healthy participants, individuals homozygous for the minor allele at rs7587026 displayed significantly reduced mean hippocampal volume compared to major allele homozygotes (Cohen's D = - 0.28, P = 0.02), and to heterozygotes (Cohen's D = - 0.36, P = 0.009). Compared to wild type, scn1lab-overexpressing zebrafish larvae exhibited more frequent spontaneous seizures [one-way ANOVA F(4,54) = 6.95 (P < 0.001)]. The number of EEG discharges correlated with the level of scn1lab overexpression [one-way ANOVA F(4,15) = 10.75 (P < 0.001]. Finally, we showed that a 50 bp promoter motif containing rs7587026 exerts a strong regulatory role on SCN1A expression, though we could not directly link this to rs7587026 itself. Our results develop the mechanistic link between rs7587026 and mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures. Furthermore, we propose that quantitative precision may be important when increasing SCN1A expression in current strategies aiming to treat seizures in conditions involving SCN1A haploinsufficiency, such as Dravet syndrome.
Collapse
|
10
|
Müller JA, Betzin J, Santos-Tejedor J, Mayer A, Oprişoreanu AM, Engholm-Keller K, Paulußen I, Gulakova P, McGovern TD, Gschossman LJ, Schönhense E, Wark JR, Lamprecht A, Becker AJ, Waardenberg AJ, Graham ME, Dietrich D, Schoch S. A presynaptic phosphosignaling hub for lasting homeostatic plasticity. Cell Rep 2022; 39:110696. [PMID: 35443170 DOI: 10.1016/j.celrep.2022.110696] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/26/2021] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Abstract
Stable function of networks requires that synapses adapt their strength to levels of neuronal activity, and failure to do so results in cognitive disorders. How such homeostatic regulation may be implemented in mammalian synapses remains poorly understood. Here we show that the phosphorylation status of several positions of the active-zone (AZ) protein RIM1 are relevant for synaptic glutamate release. Position RIMS1045 is necessary and sufficient for expression of silencing-induced homeostatic plasticity and is kept phosphorylated by serine arginine protein kinase 2 (SRPK2). SRPK2-induced upscaling of synaptic release leads to additional RIM1 nanoclusters and docked vesicles at the AZ and is not observed in the absence of RIM1 and occluded by RIMS1045E. Our data suggest that SRPK2 and RIM1 represent a presynaptic phosphosignaling hub that is involved in the homeostatic balance of synaptic coupling of neuronal networks.
Collapse
Affiliation(s)
- Johannes Alexander Müller
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Julia Betzin
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Jorge Santos-Tejedor
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Annika Mayer
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Ana-Maria Oprişoreanu
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Kasper Engholm-Keller
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark; Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | | | - Polina Gulakova
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | | | - Lena Johanna Gschossman
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Eva Schönhense
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Jesse R Wark
- Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Alf Lamprecht
- Department of Pharmaceutics, Bonn University, Bonn, Germany
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Ashley J Waardenberg
- Australian Institute for Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878, Australia; i-Synapse, Cairns, QLD, Australia
| | - Mark E Graham
- Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Dirk Dietrich
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany.
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Felix R, Muñoz-Herrera D, Corzo-López A, Fernández-Gallardo M, Leyva-Leyva M, González-Ramírez R, Sandoval A. Ion channel long non-coding RNAs in neuropathic pain. Pflugers Arch 2022; 474:457-468. [PMID: 35235008 DOI: 10.1007/s00424-022-02675-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023]
Abstract
Neuropathic pain is one of the primary forms of chronic pain and is the consequence of the somatosensory system's direct injury or disease. It is a relevant public health problem that affects about 10% of the world's general population. In neuropathic pain, alteration in neurotransmission occurs at various levels, including the dorsal root ganglia, the spinal cord, and the brain, resulting from the malfunction of diverse molecules such as receptors, ion channels, and elements of specific intracellular signaling pathways. In this context, there have been exciting advances in elucidating neuropathic pain's cellular and molecular mechanisms in the last decade, including the possible role that long non-coding RNAs (lncRNAs) may play, which open up new alternatives for the development of diagnostic and therapeutic strategies for this condition. This review focuses on recent studies associated with the possible relevance of lncRNAs in the development and maintenance of neuropathic pain through their actions on the functional expression of ion channels. Recognizing the changes in the function and spatio-temporal patterns of expression of these membrane proteins is crucial to understanding the control of neuronal excitability in chronic pain syndromes.
Collapse
Affiliation(s)
- Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico.
| | - David Muñoz-Herrera
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | | | - Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| |
Collapse
|
12
|
Tsortouktzidis D, Tröscher AR, Schulz H, Opitz T, Schoch S, Becker AJ, van Loo KMJ. A Versatile Clustered Regularly Interspaced Palindromic Repeats Toolbox to Study Neurological CaV3.2 Channelopathies by Promoter-Mediated Transcription Control. Front Mol Neurosci 2022; 14:667143. [PMID: 35069110 PMCID: PMC8770422 DOI: 10.3389/fnmol.2021.667143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 12/15/2021] [Indexed: 11/15/2022] Open
Abstract
Precise genome editing in combination with viral delivery systems provides a valuable tool for neuroscience research. Traditionally, the role of genes in neuronal circuits has been addressed by overexpression or knock-out/knock-down systems. However, those techniques do not manipulate the endogenous loci and therefore have limitations. Those constraints include that many genes exhibit extensive alternative splicing, which can be regulated by neuronal activity. This complexity cannot be easily reproduced by overexpression of one protein variant. The CRISPR activation and interference/inhibition systems (CRISPRa/i) directed to promoter sequences can modulate the expression of selected target genes in a highly specific manner. This strategy could be particularly useful for the overexpression of large proteins and for alternatively spliced genes, e.g., for studying large ion channels known to be affected in ion channelopathies in a variety of neurological diseases. Here, we demonstrate the feasibility of a newly developed CRISPRa/i toolbox to manipulate the promoter activity of the Cacna1h gene. Impaired, function of the low-voltage-activated T-Type calcium channel CaV3.2 is involved in genetic/mutational as well as acquired/transcriptional channelopathies that emerge with epileptic seizures. We show CRISPR-induced activation and inhibition of the Cacna1h locus in NS20Y cells and primary cortical neurons, as well as activation in mouse organotypic slice cultures. In future applications, the system offers the intriguing perspective to study functional effects of gain-of-function or loss-of-function variations in the Cacna1h gene in more detail. A better understanding of CaV3.2 channelopathies might result in a major advancement in the pharmacotherapy of CaV3.2 channelopathy diseases.
Collapse
Affiliation(s)
- Despina Tsortouktzidis
- Institute of Neuropathology, Medical Faculty, Section for Translational Epilepsy Research, University of Bonn, Bonn, Germany
| | - Anna R. Tröscher
- Institute of Neuropathology, Medical Faculty, Section for Translational Epilepsy Research, University of Bonn, Bonn, Germany
- Department of Neurology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany
| | - Thoralf Opitz
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Susanne Schoch
- Institute of Neuropathology, Medical Faculty, Section for Translational Epilepsy Research, University of Bonn, Bonn, Germany
| | - Albert J. Becker
- Institute of Neuropathology, Medical Faculty, Section for Translational Epilepsy Research, University of Bonn, Bonn, Germany
| | - Karen M. J. van Loo
- Institute of Neuropathology, Medical Faculty, Section for Translational Epilepsy Research, University of Bonn, Bonn, Germany
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany
- *Correspondence: Karen M. J. van Loo,
| |
Collapse
|
13
|
Ste20-like Kinase Is Critical for Inhibitory Synapse Maintenance and Its Deficiency Confers a Developmental Dendritopathy. J Neurosci 2021; 41:8111-8125. [PMID: 34400520 DOI: 10.1523/jneurosci.0352-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/18/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
The size and structure of the dendritic arbor play important roles in determining how synaptic inputs of neurons are converted to action potential output. The regulatory mechanisms governing the development of dendrites, however, are insufficiently understood. The evolutionary conserved Ste20/Hippo kinase pathway has been proposed to play an important role in regulating the formation and maintenance of dendritic architecture. A key element of this pathway, Ste20-like kinase (SLK), regulates cytoskeletal dynamics in non-neuronal cells and is strongly expressed throughout neuronal development. However, its function in neurons is unknown. We show that, during development of mouse cortical neurons, SLK has a surprisingly specific role for proper elaboration of higher, ≥ third-order dendrites both in male and in female mice. Moreover, we demonstrate that SLK is required to maintain excitation-inhibition balance. Specifically, SLK knockdown caused a selective loss of inhibitory synapses and functional inhibition after postnatal day 15, whereas excitatory neurotransmission was unaffected. Finally, we show that this mechanism may be relevant for human disease, as dysmorphic neurons within human cortical malformations revealed significant loss of SLK expression. Overall, the present data identify SLK as a key regulator of both dendritic complexity during development and inhibitory synapse maintenance.SIGNIFICANCE STATEMENT We show that dysmorphic neurons of human epileptogenic brain lesions have decreased levels of the Ste20-like kinase (SLK). Decreasing SLK expression in mouse neurons revealed that SLK has essential functions in forming the neuronal dendritic tree and in maintaining inhibitory connections with neighboring neurons.
Collapse
|
14
|
Targeting T-type/CaV3.2 channels for chronic pain. Transl Res 2021; 234:20-30. [PMID: 33422652 PMCID: PMC8217081 DOI: 10.1016/j.trsl.2021.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 01/09/2023]
Abstract
T-type calcium channels regulate neuronal excitability and are important contributors of pain processing. CaV3.2 channels are the major isoform expressed in nonpeptidergic and peptidergic nociceptive neurons and are emerging as promising targets for pain treatment. Numerous studies have shown that CaV3.2 expression and/or activity are significantly increased in spinal dorsal horn and in dorsal root ganglia neurons in different inflammatory and neuropathic pain models. Pharmacological campaigns to inhibit the functional expression of CaV3.2 for treatment of pain have focused on the development of direct channel blockers, but none have produced lead candidates. Targeting the proteins that regulate the trafficking or transcription, and the ones that modify the channels via post-translational modifications are alternative means to regulate expression and function of CaV3.2 channels and hence to develop new drugs to control pain. Here we synthesize data supporting a role for CaV3.2 in numerous pain modalities and then discuss emerging opportunities for the indirect targeting of CaV3.2 channels.
Collapse
|
15
|
Buffolo F, Petrosino V, Albini M, Moschetta M, Carlini F, Floss T, Kerlero de Rosbo N, Cesca F, Rocchi A, Uccelli A, Benfenati F. Neuroinflammation induces synaptic scaling through IL-1β-mediated activation of the transcriptional repressor REST/NRSF. Cell Death Dis 2021; 12:180. [PMID: 33589593 PMCID: PMC7884694 DOI: 10.1038/s41419-021-03465-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is associated with synapse dysfunction and cognitive decline in patients and animal models. One candidate for translating the inflammatory stress into structural and functional changes in neural networks is the transcriptional repressor RE1-silencing transcription factor (REST) that regulates the expression of a wide cluster of neuron-specific genes during neurogenesis and in mature neurons. To study the cellular and molecular pathways activated under inflammatory conditions mimicking the experimental autoimmune encephalomyelitis (EAE) environment, we analyzed REST activity in neuroblastoma cells and mouse cortical neurons treated with activated T cell or microglia supernatant and distinct pro-inflammatory cytokines. We found that REST is activated by a variety of neuroinflammatory stimuli in both neuroblastoma cells and primary neurons, indicating that a vast transcriptional change is triggered during neuroinflammation. While a dual activation of REST and its dominant-negative splicing isoform REST4 was observed in N2a neuroblastoma cells, primary neurons responded with a pure full-length REST upregulation in the absence of changes in REST4 expression. In both cases, REST upregulation was associated with activation of Wnt signaling and increased nuclear translocation of β-catenin, a well-known intracellular transduction pathway in neuroinflammation. Among single cytokines, IL-1β caused a potent and prompt increase in REST transcription and translation in neurons, which promoted a delayed and strong synaptic downscaling specific for excitatory synapses, with decreased frequency and amplitude of spontaneous synaptic currents, decreased density of excitatory synaptic connections, and decreased frequency of action potential-evoked Ca2+ transients. Most important, the IL-1β effects on excitatory transmission were strictly REST dependent, as conditional deletion of REST completely occluded the effects of IL-1β activation on synaptic transmission and network excitability. Our results demonstrate that REST upregulation represents a new pathogenic mechanism for the synaptic dysfunctions observed under neuroinflammatory conditions and identify the REST pathway as therapeutic target for EAE and, potentially, for multiple sclerosis.
Collapse
Affiliation(s)
- Federica Buffolo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132, Genova, Italy
| | - Valentina Petrosino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo P. Daneo, 3, 16132, Genova, Italy
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
| | - Martina Albini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132, Genova, Italy
| | - Matteo Moschetta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
| | - Federico Carlini
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo P. Daneo, 3, 16132, Genova, Italy
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
| | - Thomas Floss
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Nicole Kerlero de Rosbo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo P. Daneo, 3, 16132, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Anna Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy.
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo P. Daneo, 3, 16132, Genova, Italy.
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy.
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| |
Collapse
|
16
|
Tsortouktzidis D, Schulz H, Hamed M, Vatter H, Surges R, Schoch S, Sander T, Becker AJ, van Loo KMJ. Gene expression analysis in epileptic hippocampi reveals a promoter haplotype conferring reduced aldehyde dehydrogenase 5a1 expression and responsiveness. Epilepsia 2020; 62:e29-e34. [PMID: 33319393 DOI: 10.1111/epi.16789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/22/2020] [Accepted: 11/22/2020] [Indexed: 11/29/2022]
Abstract
Increasing evidence indicates the pathogenetic relevance of regulatory genomic motifs for variability in the manifestation of brain disorders. In this context, cis-regulatory effects of single nucleotide polymorphisms (SNPs) on gene expression can contribute to changing transcript levels of excitability-relevant molecules and episodic seizure manifestation in epilepsy. Biopsy specimens of patients undergoing epilepsy surgery for seizure relief provide unique insights into the impact of promoter SNPs on corresponding mRNA expression. Here, we have scrutinized whether two linked regulatory SNPs (rs2744575; 4779C > G and rs4646830; 4854C > G) located in the aldehyde dehydrogenase 5a1 (succinic semialdehyde dehydrogenase; ALDH5A1) gene promoter are associated with expression of corresponding mRNAs in epileptic hippocampi (n = 43). The minor ALDH5A1-GG haplotype associates with significantly lower ALDH5A1 transcript abundance. Complementary in vitro analyses in neural cell cultures confirm this difference and further reveal a significantly constricted range for the minor ALDH5A1 haplotype of promoter activity regulation through the key epileptogenesis transcription factor Egr1 (early growth response 1). The present data suggest systematic analyses in human hippocampal tissue as a useful approach to unravel the impact of epilepsy candidate SNPs on associated gene expression. Aberrant ALDH5A1 promoter regulation in functional terms can contribute to impaired γ-aminobutyric acid homeostasis and thereby network excitability and seizure propensity.
Collapse
Affiliation(s)
- Despina Tsortouktzidis
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Herbert Schulz
- Cologne Center of Genomics, University of Cologne, Germany
| | - Motaz Hamed
- Clinic for Neurosurgery, University of Bonn Medical Center, Bonn, Germany
| | - Hartmut Vatter
- Clinic for Neurosurgery, University of Bonn Medical Center, Bonn, Germany
| | - Rainer Surges
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Thomas Sander
- Cologne Center of Genomics, University of Cologne, Germany
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Karen M J van Loo
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany.,Department of Epileptology, Neurology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
17
|
Tomita S, Sekiguchi F, Kasanami Y, Naoe K, Tsubota M, Wake H, Nishibori M, Kawabata A. Ca v3.2 overexpression in L4 dorsal root ganglion neurons after L5 spinal nerve cutting involves Egr-1, USP5 and HMGB1 in rats: An emerging signaling pathway for neuropathic pain. Eur J Pharmacol 2020; 888:173587. [PMID: 32971090 DOI: 10.1016/j.ejphar.2020.173587] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Overexpression of Cav3.2 T-type Ca2+ channels in L4 dorsal root ganglion (DRG) participates in neuropathic pain after L5 spinal nerve cutting (L5SNC) in rats. The L5SNC-induced neuropathic pain also involves high mobility group box 1 (HMGB1), a damage-associated molecular pattern protein, and its target, the receptor for advanced glycation end-products (RAGE). We thus studied the molecular mechanisms for the L5SNC-induced Cav3.2 overexpression as well as neuropathic pain in rats by focusing on; 1) possible involvement of early growth response 1 (Egr-1), known to regulate transcriptional expression of Cav3.2, and ubiquitin-specific protease 5 (USP5) that protects Cav3.2 from proteasomal degradation, and 2) possible role of HMGB1/RAGE as an upstream signal. Protein levels of Cav3.2 as well as Egr-1 in L4 DRG significantly increased in the early (day 6) and persistent (day 14) phases of neuropathy after L5SNC, while USP5 protein in L4 DRG did not increase on day 6, but day 14. An anti-HMGB1-neutralizing antibody or a low molecular weight heparin, a RAGE antagonist, prevented the development of neuropathic pain and upregulation of Egr-1 and Cav3.2 in L4 DRG after L5SNC. L5SNC increased macrophages accumulating in the sciatic nerves, and the cytoplasm/nuclear ratio of immunoreactive HMGB1 in those macrophages. Our findings suggest that L5SNC-induced Cav3.2 overexpression in L4 DRG and neuropathic pain involves Egr-1 upregulation downstream of the macrophage-derived HMGB1/RAGE pathway, and that the delayed upregulation of USP5 might contribute to the persistent Cav3.2 overexpression and neuropathy.
Collapse
Affiliation(s)
- Shiori Tomita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yoshihito Kasanami
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Katsuki Naoe
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
18
|
Thalhammer A, Jaudon F, Cingolani LA. Emerging Roles of Activity-Dependent Alternative Splicing in Homeostatic Plasticity. Front Cell Neurosci 2020; 14:104. [PMID: 32477067 PMCID: PMC7235277 DOI: 10.3389/fncel.2020.00104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022] Open
Abstract
Homeostatic plasticity refers to the ability of neuronal networks to stabilize their activity in the face of external perturbations. Most forms of homeostatic plasticity ultimately depend on changes in the expression or activity of ion channels and synaptic proteins, which may occur at the gene, transcript, or protein level. The most extensively investigated homeostatic mechanisms entail adaptations in protein function or localization following activity-dependent posttranslational modifications. Numerous studies have also highlighted how homeostatic plasticity can be achieved by adjusting local protein translation at synapses or transcription of specific genes in the nucleus. In comparison, little attention has been devoted to whether and how alternative splicing (AS) of pre-mRNAs underlies some forms of homeostatic plasticity. AS not only expands proteome diversity but also contributes to the spatiotemporal dynamics of mRNA transcripts. Prominent in the brain where it can be regulated by neuronal activity, it is a flexible process, tightly controlled by a multitude of factors. Given its extensive use and versatility in optimizing the function of ion channels and synaptic proteins, we argue that AS is ideally suited to achieve homeostatic control of neuronal output. We support this thesis by reviewing emerging evidence linking AS to various forms of homeostatic plasticity: homeostatic intrinsic plasticity, synaptic scaling, and presynaptic homeostatic plasticity. Further, we highlight the relevance of this connection for brain pathologies.
Collapse
Affiliation(s)
- Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
19
|
|
20
|
Sholler DJ, Merritt CR, Davis-Reyes BD, Golovko G, Anastasio NC, Cunningham KA. Inherent Motor Impulsivity Associates with Specific Gene Targets in the Rat Medial Prefrontal Cortex. Neuroscience 2020; 435:161-173. [PMID: 32240784 DOI: 10.1016/j.neuroscience.2020.03.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/22/2020] [Accepted: 03/26/2020] [Indexed: 01/23/2023]
Abstract
High impulsivity characterizes a myriad of neuropsychiatric diseases, and identifying targets for neuropharmacological intervention to reduce impulsivity could reveal transdiagnostic treatment strategies. Motor impulsivity (impulsive action) reflects in part the failure of "top-down" executive control by the medial prefrontal cortex (mPFC). The present study profiled the complete set of mRNA molecules expressed from genes (transcriptome) in the mPFC of male, outbred rats stably expressing high (HI) or low (LI) motor impulsivity based upon premature responses in the 1-choice serial reaction time (1-CSRT) task. RNA-sequencing identified expression of 18 genes that was higher in the mPFC of HI vs. LI rats. Functional gene enrichment revealed that biological processes related to calcium homeostasis and G protein-coupled receptor (GPCR) signaling pathways, particularly glutamatergic, were overrepresented in the mPFC of HI vs. LI rats. Transcription factor enrichment identified mothers against decapentaplegic homolog 4 (SMAD4) and RE1 silencing transcription factor (REST) as overrepresented in the mPFC of HI rats relative to LI rats, while in silico analysis predicted a conserved SMAD binding site within the voltage-gated calcium channel subunit alpha1 E (CACNA1E) promoter region. qRT-PCR analyses confirmed that mRNA expression of CACNA1E, as well as expression of leucyl and cystinyl aminopeptidase (LNPEP), were higher in the mPFC of HI vs. LI rats. These outcomes establish a transcriptomic landscape in the mPFC that is related to individual differences in motor impulsivity and propose novel gene targets for future impulsivity research.
Collapse
Affiliation(s)
- Dennis J Sholler
- Center for Addiction Research, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Christina R Merritt
- Center for Addiction Research, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Brionna D Davis-Reyes
- Center for Addiction Research, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - George Golovko
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Noelle C Anastasio
- Center for Addiction Research, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Kathryn A Cunningham
- Center for Addiction Research, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| |
Collapse
|
21
|
van Loo KMJ, Becker AJ. Transcriptional Regulation of Channelopathies in Genetic and Acquired Epilepsies. Front Cell Neurosci 2020; 13:587. [PMID: 31992970 PMCID: PMC6971179 DOI: 10.3389/fncel.2019.00587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/23/2019] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a common neurological disorder characterized by recurrent uncontrolled seizures and has an idiopathic “genetic” etiology or a symptomatic “acquired” component. Genetic studies have revealed that many epilepsy susceptibility genes encode ion channels, including voltage-gated sodium, potassium and calcium channels. The high prevalence of ion channels in epilepsy pathogenesis led to the causative concept of “ion channelopathies,” which can be elicited by specific mutations in the coding or promoter regions of genes in genetic epilepsies. Intriguingly, expression changes of the same ion channel genes by augmentation of specific transcription factors (TFs) early after an insult can underlie acquired epilepsies. In this study, we review how the transcriptional regulation of ion channels in both genetic and acquired epilepsies can be controlled, and compare these epilepsy “ion channelopathies” with other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Karen M J van Loo
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, Bonn, Germany
| | - Albert J Becker
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
22
|
Carminati E, Buffolo F, Rocchi A, Michetti C, Cesca F, Benfenati F. Mild Inactivation of RE-1 Silencing Transcription Factor (REST) Reduces Susceptibility to Kainic Acid-Induced Seizures. Front Cell Neurosci 2020; 13:580. [PMID: 31998079 PMCID: PMC6965066 DOI: 10.3389/fncel.2019.00580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/18/2019] [Indexed: 11/13/2022] Open
Abstract
RE-1 Silencing Transcription factor (REST) controls several steps in neural development by modulating the expression of a wide range of neural genes. Alterations in REST expression have been associated with the onset of epilepsy; however, whether such alterations are deleterious or represent a protective homeostatic response remains elusive. To study the impact of REST modulation on seizure propensity, we developed a tool for its negative modulation in vivo. The tool is composed of the paired-amphipathic helix 1 (PAH1) domain, a competitive inhibitor of REST activation by mSin3, fused to the light-oxygen-voltage sensing 2 (LOV2) domain of Avena sativa phototropin 1, a molecular switch to alternatively hide or expose the PAH1 inhibitor. We employed the C450A and I539E light-independent AsLOV2 variants to mimic the closed (inactive) and open (active) states of LOV2-PAH1, respectively. Recombinant AAV1/2 viral particles (rAAVs) allowed LOV2-PAH1 expression in HEK293T cells and primary neurons, and efficiently transduced hippocampal neurons in vivo. mRNA expression analysis revealed an increased expression of several neuronal genes in the hippocampi of mice expressing the open probe. AAV-transduced mice received a single dose of kainic acid (KA), a treatment known to induce a transient increase of REST levels in the hippocampus. Remarkably, mice expressing the active variant displayed a reduced number of KA-induced seizures, which were less severe compared to mice carrying the inactive probe. These data support the validity of our tool to modulate REST activity in vivo and the potential impact of REST modulation on epileptogenesis.
Collapse
Affiliation(s)
- Emanuele Carminati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Federica Buffolo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Anna Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
23
|
Sekiguchi F, Kawabata A. [Role of Ca v3.2 T-type Ca 2+ channels in prostate cancer cells]. Nihon Yakurigaku Zasshi 2019; 154:97-102. [PMID: 31527367 DOI: 10.1254/fpj.154.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Among voltage-gated Ca2+ channels, T-type Ca2+ channels, which are activated by low voltages, regulate neuronal excitability, spontaneous neurotransmitter release, hormone secretion, etc. and also participate in proliferation of distinct cancer cells. Among three isoforms of T-type Ca2+ channels, Cav3.2 is detectable in 100% of biopsy samples from prostate cancer patients. In general, prostate cancer cells are highly sensitive to androgen deprivation therapy, but often acquire hormone-therapy resistance. The androgen deprivation may trigger neuroendocrine (NE)-like differentiation of some prostate cancer cells. We have analyzed the expression and function of Cav3.2 in human prostate cancer LNCaP cells during NE-like differentiation. NE-like LNCaP cells overexpress Cav3.2 through the CREB/Egr-1 pathway and also cystathionine-γ-lyase (CSE), which generates H2S that enhances the channel activity of Cav3.2. H2S generated by upregulated CSE appears to enhance the activity of upregulated Cav3.2 after the differentiation. The enhanced Cav3.2 activity in NE-like cells may contribute to increased secretion of mitogenic factors essential for androgen-independent proliferation of surrounding prostate cancer cells. It is known that increased extracellular glucose levels enhance Cav3.2 activity through asparagine (N)-linked glycosylation of Cav3.2, which might contribute to diabetic neuropathy. We then found that high glucose accelerates the enhanced channel function and overexpression of Cav3.2 in NE-like LNCaP cells, which might be associated with clinical evidence for diabetes-related poor prognosis of prostate cancer and development of hormone therapy resistance. Thus, Cav3.2 is considered to play a role in the pathophysiology of prostate cancer, and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| |
Collapse
|
24
|
Cav3.2 T-type calcium channels shape electrical firing in mouse Lamina II neurons. Sci Rep 2019; 9:3112. [PMID: 30816223 PMCID: PMC6395820 DOI: 10.1038/s41598-019-39703-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
The T-type calcium channel, Cav3.2, is necessary for acute pain perception, as well as mechanical and cold allodynia in mice. Being found throughout sensory pathways, from excitatory primary afferent neurons up to pain matrix structures, it is a promising target for analgesics. In our study, Cav3.2 was detected in ~60% of the lamina II (LII) neurons of the spinal cord, a site for integration of sensory processing. It was co-expressed with Tlx3 and Pax2, markers of excitatory and inhibitory interneurons, as well as nNOS, calretinin, calbindin, PKCγ and not parvalbumin. Non-selective T-type channel blockers slowed the inhibitory but not the excitatory transmission in LII neurons. Furthermore, T-type channel blockers modified the intrinsic properties of LII neurons, abolishing low-threshold activated currents, rebound depolarizations, and blunting excitability. The recording of Cav3.2-positive LII neurons, after intraspinal injection of AAV-DJ-Cav3.2-mcherry, showed that their intrinsic properties resembled those of the global population. However, Cav3.2 ablation in the dorsal horn of Cav3.2GFP-Flox KI mice after intraspinal injection of AAV-DJ-Cav3.2-Cre-IRES-mcherry, had drastic effects. Indeed, it (1) blunted the likelihood of transient firing patterns; (2) blunted the likelihood and the amplitude of rebound depolarizations, (3) eliminated action potential pairing, and (4) remodeled the kinetics of the action potentials. In contrast, the properties of Cav3.2-positive neurons were only marginally modified in Cav3.1 knockout mice. Overall, in addition to their previously established roles in the superficial spinal cord and in primary afferent neurons, Cav3.2 channel appear to be necessary for specific, significant and multiple controls of LII neuron excitability.
Collapse
|
25
|
Calcium Channel Subunit α2δ4 Is Regulated by Early Growth Response 1 and Facilitates Epileptogenesis. J Neurosci 2019; 39:3175-3187. [PMID: 30792272 DOI: 10.1523/jneurosci.1731-18.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/03/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
Transient brain insults, including status epilepticus (SE), can trigger a period of epileptogenesis during which functional and structural reorganization of neuronal networks occurs resulting in the onset of focal epileptic seizures. In recent years, mechanisms that regulate the dynamic transcription of individual genes during epileptogenesis and thereby contribute to the development of a hyperexcitable neuronal network have been elucidated. Our own results have shown early growth response 1 (Egr1) to transiently increase expression of the T-type voltage-dependent Ca2+ channel (VDCC) subunit CaV3.2, a key proepileptogenic protein. However, epileptogenesis involves complex and dynamic transcriptomic alterations; and so far, our understanding of the transcriptional control mechanism of gene regulatory networks that act in the same processes is limited. Here, we have analyzed whether Egr1 acts as a key transcriptional regulator for genes contributing to the development of hyperexcitability during epileptogenesis. We found Egr1 to drive the expression of the VDCC subunit α2δ4, which was augmented early and persistently after pilocarpine-induced SE. Furthermore, we show that increasing levels of α2δ4 in the CA1 region of the hippocampus elevate seizure susceptibility of mice by slightly decreasing local network activity. Interestingly, we also detected increased expression levels of Egr1 and α2δ4 in human hippocampal biopsies obtained from epilepsy surgery. In conclusion, Egr1 controls the abundance of the VDCC subunits CaV3.2 and α2δ4, which act synergistically in epileptogenesis, and thereby contributes to a seizure-induced "transcriptional Ca2+ channelopathy."SIGNIFICANCE STATEMENT The onset of focal recurrent seizures often occurs after an epileptogenic process induced by transient insults to the brain. Recently, transcriptional control mechanisms for individual genes involved in converting neurons hyperexcitable have been identified, including early growth response 1 (Egr1), which activates transcription of the T-type Ca2+ channel subunit CaV3.2. Here, we find Egr1 to regulate also the expression of the voltage-dependent Ca2+ channel subunit α2δ4, which was augmented after pilocarpine- and kainic acid-induced status epilepticus. In addition, we observed that α2δ4 affected spontaneous network activity and the susceptibility for seizure induction. Furthermore, we detected corresponding dynamics in human biopsies from epilepsy patients. In conclusion, Egr1 orchestrates a seizure-induced "transcriptional Ca2+ channelopathy" consisting of CaV3.2 and α2δ4, which act synergistically in epileptogenesis.
Collapse
|
26
|
T-type Calcium Channels in Cancer. Cancers (Basel) 2019; 11:cancers11020134. [PMID: 30678110 PMCID: PMC6407089 DOI: 10.3390/cancers11020134] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Although voltage-activated Ca2+ channels are a common feature in excitable cells, their expression in cancer tissue is less understood. T-type Ca2+ channels are particularly overexpressed in various cancers. Because of their activation profile at membrane potentials close to rest and the generation of a window current, T-type Ca2+ channels may regulate a variety of Ca2+-dependent cellular processes, including cell proliferation, survival, and differentiation. The expression of T-type Ca2+ channels is of special interest as a target for therapeutic interventions.
Collapse
|
27
|
Tomita S, Sekiguchi F, Deguchi T, Miyazaki T, Ikeda Y, Tsubota M, Yoshida S, Nguyen HD, Okada T, Toyooka N, Kawabata A. Critical role of Ca v3.2 T-type calcium channels in the peripheral neuropathy induced by bortezomib, a proteasome-inhibiting chemotherapeutic agent, in mice. Toxicology 2018; 413:33-39. [PMID: 30552955 DOI: 10.1016/j.tox.2018.12.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/30/2023]
Abstract
Bortezomib, a first-line agent for treatment of multiple myeloma, exhibits anticancer activity through proteasome inhibition. However, bortezomib-induced peripheral neuropathy (BIPN) is one of the most serious side effects. Since decreased proteasomal degradation of Cav3.2 T-type calcium channels in the primary afferents is involved in persistent pain, we investigated whether BIPN involves increased protein levels of Cav3.2 in mice. Six repeated i.p. administrations of bortezomib for 12 days developed persistent mechanical allodynia. Systemic administration of novel T-type calcium channel blockers, (2R/S)-6-prenylnaringenin and KTt-45, and of TTA-A2, the well-known blocker, reversed the BIPN. Ascorbic acid, known to block Cav3.2, but not Cav3.1 or 3.3, and silencing of Cav3.2 gene also suppressed BIPN. Protein levels of Cav3.2 in the dorsal root ganglion (DRG) at L4-L6 levels increased throughout days 1-21 after the onset of bortezomib treatment. Protein levels of USP5, a deubiquitinating enzyme that specifically inhibits proteasomal degradation of Cav3.2, increased in DRG on days 3-21, but not day 1, in bortezomib-treated mice. In DRG-derived ND7/23 cells, bortezomib increased protein levels of Cav3.2 and T-channel-dependent currents, as assessed by a patch-clamp method, but did not upregulate expression of Cav3.2 mRNA or USP5 protein. MG-132, another proteasome inhibitor, also increased Cav3.2 protein levels in the cultured cells. Given the previous evidence for USP5 induction following nociceptor excitation, our data suggest that BIPN involves the increased protein levels of Cav3.2 in nociceptors through inhibition of proteasomal degradation of Cav3.2 by bortezomib itself and then by USP5 that is upregulated probably in an activity-dependent manner.
Collapse
Affiliation(s)
- Shiori Tomita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Tomoyo Deguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Takaya Miyazaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Yuya Ikeda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Huy Du Nguyen
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan; Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan.
| |
Collapse
|
28
|
Falcón D, González-Montelongo R, Sánchez de Rojas-de Pedro E, Ordóñez A, Ureña J, Castellano A. Dexamethasone-induced upregulation of Ca V3.2 T-type Ca 2+ channels in rat cardiac myocytes. J Steroid Biochem Mol Biol 2018; 178:193-202. [PMID: 29262379 DOI: 10.1016/j.jsbmb.2017.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 11/21/2017] [Accepted: 12/14/2017] [Indexed: 12/27/2022]
Abstract
Glucocorticoids are widely used to treat acute and chronic diseases. Unfortunately, their therapeutic use is associated with severe side effects. Glucocorticoids are known to regulate several ion channels in cardiac myocytes, including voltage-dependent Ca2+ channels. Low-voltage-activated T-type Ca2+ channels are expressed in ventricular myocytes during the fetal and perinatal period, but are practically absent in the adult. However, these channels can be re-expressed in adult cardiomyocytes under some pathological conditions. We have investigated the glucocorticoid regulation of T-type Ca2+ channels in rat cardiomyocytes. Molecular studies revealed that dexamethasone induces the upregulation of CaV3.2 mRNA in neonatal rat ventricular myocytes, whereas CaV3.1 mRNA is only slightly affected. Patch-clamp recordings confirmed that T-type Ca2+ channel currents were upregulated in dexamethasone treated cardiomyocytes, and the addition of 50 μmol/L NiCl2 demonstrated that the CaV3.2 channel is responsible for this upregulation. The effect of dexamethasone on CaV3.2 is mediated by the activation and translocation to the cell nucleus of the glucocorticoid receptor (GR). We have isolated the upstream promoter of the Cacna1h gene and tested its activity in transfected ventricular myocytes. The initial in silico analysis of Cacna1h promoter revealed putative glucocorticoid response elements (GREs). Transcriptional activity assays combined with deletion analyses and chromatin immunoprecipitation assays demonstrated that GR binds to a region a GRE located in -1006/-985 bp of Cacna1h promoter. Importantly, upregulation of the CaV3.2 channel is also observed in vitro in adult rat ventricular myocytes, and in vivo in a rat model of excess of glucocorticoids.
Collapse
Affiliation(s)
- D Falcón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - R González-Montelongo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - E Sánchez de Rojas-de Pedro
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain
| | - A Ordóñez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain; CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - J Ureña
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - A Castellano
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain.
| |
Collapse
|
29
|
Becker AJ. Review: Animal models of acquired epilepsy: insights into mechanisms of human epileptogenesis. Neuropathol Appl Neurobiol 2018; 44:112-129. [DOI: 10.1111/nan.12451] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/27/2017] [Indexed: 02/06/2023]
Affiliation(s)
- A. J. Becker
- Section for Translational Epilepsy Research; Department of Neuropathology; University of Bonn Medical Center; Bonn Germany
| |
Collapse
|
30
|
González-Ramírez R, Felix R. Transcriptional regulation of voltage-gated Ca 2+ channels. Acta Physiol (Oxf) 2018; 222. [PMID: 28371478 DOI: 10.1111/apha.12883] [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: 12/20/2016] [Revised: 02/14/2017] [Accepted: 03/21/2017] [Indexed: 12/30/2022]
Abstract
The transcriptional regulation of voltage-gated Ca2+ (CaV ) channels is an emerging research area that promises to improve our understanding of how many relevant physiological events are shaped in the central nervous system, the skeletal muscle and other tissues. Interestingly, a picture of how transcription of CaV channel subunit genes is controlled is evolving with the identification of the promoter regions required for tissue-specific expression and the identification of transcription factors that control their expression. These promoters share several characteristics that include multiple transcriptional start sites, lack of a TATA box and the presence of elements conferring tissue-selective expression. Likewise, changes in CaV channel expression occur throughout development, following ischaemia, seizures or chronic drug administration. This review focuses on insights achieved regarding the control of CaV channel gene expression. To further understand the complexities of expression and to increase the possibilities of detecting CaV channel alterations causing human disease, a deeper knowledge on the structure of the 5' upstream regions of the genes encoding these remarkable proteins will be necessary.
Collapse
Affiliation(s)
- R. González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad; Hospital General ‘Dr. Manuel Gea González’; Secretaría de Salud; Ciudad de México México
| | - R. Felix
- Departmento de Biología Celular; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN); Ciudad de México México
| |
Collapse
|
31
|
Zinc deficiency promotes cystitis-related bladder pain by enhancing function and expression of Ca v3.2 in mice. Toxicology 2017; 393:102-112. [PMID: 29129814 DOI: 10.1016/j.tox.2017.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/08/2017] [Accepted: 11/08/2017] [Indexed: 01/15/2023]
Abstract
Cav3.2 T-type Ca2+ channel activity is suppressed by zinc that binds to the extracellular histidine-191 of Cav3.2, and enhanced by H2S that interacts with zinc. Cav3.2 in nociceptors is upregulated in an activity-dependent manner. The enhanced Cav3.2 activity by H2S formed by the upregulated cystathionine-γ-lyase (CSE) is involved in the cyclophosphamide (CPA)-induced cystitis-related bladder pain in mice. We thus asked if zinc deficiency affects the cystitis-related bladder pain in mice by altering Cav3.2 function and/or expression. Dietary zinc deficiency for 2 weeks greatly decreased zinc concentrations in the plasma but not bladder tissue, and enhanced the bladder pain/referred hyperalgesia (BP/RH) following CPA at 200mg/kg, a subeffective dose, but not 400mg/kg, a maximal dose, an effect abolished by pharmacological blockade or gene silencing of Cav3.2. Acute zinc deficiency caused by systemic N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylendiamine (TPEN), a zinc chelator, mimicked the dietary zinc deficiency-induced Cav3.2-dependent promotion of BP/RH following CPA at 200mg/kg. CPA at 400mg/kg alone or TPEN plus CPA at 200mg/kg caused Cav3.2 overexpression accompanied by upregulation of Egr-1 and USP5, known to promote transcriptional expression and reduce proteasomal degradation of Cav3.2, respectively, in the dorsal root ganglia (DRG). The CSE inhibitor, β-cyano-l-alanine, prevented the BP/RH and upregulation of Cav3.2, Egr-1 and USP5 in DRG following TPEN plus CPA at 200mg/kg. Together, zinc deficiency promotes bladder pain accompanying CPA-induced cystitis by enhancing function and expression of Cav3.2 in nociceptors, suggesting a novel therapeutic avenue for treatment of bladder pain, such as zinc supplementation.
Collapse
|
32
|
Kostoula C, Pascente R, Ravizza T, McCown T, Schoch S, Vezzani A, Becker AJ, van Loo KMJ. Development of In Vivo Imaging Tools for Investigating Astrocyte Activation in Epileptogenesis. Mol Neurobiol 2017; 55:4463-4472. [DOI: 10.1007/s12035-017-0660-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/14/2017] [Indexed: 12/21/2022]
|
33
|
Zhang Q, Hsia SC, Martin-Caraballo M. Regulation of T-type Ca 2+ channel expression by herpes simplex virus-1 infection in sensory-like ND7 cells. J Neurovirol 2017. [PMID: 28639215 DOI: 10.1007/s13365-017-0545-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Infection of sensory neurons by herpes simplex virus (HSV)-1 disrupts electrical excitability, altering pain sensory transmission. Because of their low threshold for activation, functional expression of T-type Ca2+ channels regulates various cell functions, including neuronal excitability and neuronal communication. In this study, we have tested the effect of HSV-1 infection on the functional expression of T-type Ca2+ channels in differentiated ND7-23 sensory-like neurons. Voltage-gated Ca2+ currents were measured using whole cell patch clamp recordings in differentiated ND7-23 neurons under various culture conditions. Differentiation of ND7-23 cells evokes a significant increase in T-type Ca2+ current densities. Increased T-type Ca2+ channel expression promotes the morphological differentiation of ND7-23 cells and triggers a rebound depolarization. HSV-1 infection of differentiated ND7-23 cells causes a significant loss of T-type Ca2+ channels from the membrane. HSV-1 evoked reduction in the functional expression of T-type Ca2+ channels is mediated by several factors, including decreased expression of Cav3.2 T-type Ca2+ channel subunits and disruption of endocytic transport. Decreased functional expression of T-type Ca2+ channels by HSV-1 infection requires protein synthesis and viral replication, but occurs independently of Egr-1 expression. These findings suggest that infection of neuron-like cells by HSV-1 causes a significant disruption in the expression of T-type Ca2+ channels, which can results in morphological and functional changes in electrical excitability.
Collapse
Affiliation(s)
- Qiaojuan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, USA
| | - Shao-Chung Hsia
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, USA
| | - Miguel Martin-Caraballo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, USA.
| |
Collapse
|
34
|
Pitsch J, Becker AJ, Schoch S, Müller JA, de Curtis M, Gnatkovsky V. Circadian clustering of spontaneous epileptic seizures emerges after pilocarpine-induced status epilepticus. Epilepsia 2017; 58:1159-1171. [DOI: 10.1111/epi.13795] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Julika Pitsch
- Section for Translational Epilepsy Research; Department of Neuropathology; University of Bonn; Bonn Germany
| | - Albert J. Becker
- Section for Translational Epilepsy Research; Department of Neuropathology; University of Bonn; Bonn Germany
| | - Susanne Schoch
- Section for Translational Epilepsy Research; Department of Neuropathology; University of Bonn; Bonn Germany
| | - Johannes Alexander Müller
- Section for Translational Epilepsy Research; Department of Neuropathology; University of Bonn; Bonn Germany
| | - Marco de Curtis
- Unit of Epileptology and Experimental Neurophysiology; Fondazione Istituto Neurologico Carlo Besta; Milan Italy
| | - Vadym Gnatkovsky
- Unit of Epileptology and Experimental Neurophysiology; Fondazione Istituto Neurologico Carlo Besta; Milan Italy
| |
Collapse
|
35
|
Identification of Synaptotagmin 10 as Effector of NPAS4-Mediated Protection from Excitotoxic Neurodegeneration. J Neurosci 2016; 36:2561-70. [PMID: 26936998 DOI: 10.1523/jneurosci.2027-15.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Neuronal degeneration represents a pathogenetic hallmark after different brain insults, such as ischemia and status epilepticus (SE). Excessive release of glutamate triggered by pathophysiologic synaptic activity has been put forward as key mechanism in this context. In response to pathophysiologic synaptic activity, multiple signaling cascades are activated that ultimately initiate expression of specific sets of genes, which may decide between neuronal survival versus death. Recently, a core set of genes ["activity-regulated inhibitor of death" (AID) genes] including the transcription factor (TF) NPAS4 (neuronal PAS domain protein 4) has been found to provide activity-induced protection against neuronal death caused by excitotoxic stimulation. However, the downstream targets of AID action mediating neuroprotection remained so far unknown. Here, we have identified synaptotagmin 10 (Syt10), a vesicular Ca(2+) sensor, as the first neuroprotective effector protein downstream of the TF NPAS4. The expression of Syt10 is strongly upregulated by pathophysiologic synaptic activity after kainic acid (KA) exposure and its absence renders mouse hippocampal neurons highly susceptible to excitotoxic insults. We found NPAS4 as critical for the increase in Syt10 levels and in turn the ability of NPAS4 to confer neuroprotection against KA-induced excitotoxicity to be severely diminished in Syt10 knock-out neurons. In summary, our results point to an important role for signaling of the NPAS4-Syt10 pathway in the neuronal response to strong synaptic activity as a consequence of excitotoxic insults. SIGNIFICANCE STATEMENT Aberrant synaptic activity is observed in many neurological disorders and has been suggested as an important factor contributing to the pathophysiology. Intriguingly, pathophysiologic activity can also trigger signaling cascades mediating potentially compensatory neuroprotection against excitotoxic insult. Here, we identify a new neuroprotective signaling cascade involving the activity-induced transcriptional regulator NPAS4 and the vesicular Ca(2+)-sensor protein synaptotagmin 10 (Syt10). Syt10 is required for NPAS4 to protect hippocampal neurons against excitotoxic cell death. NPAS4 in turn controls the activity of the Syt10 gene, which is strongly induced by pathophysiologic activity. Our results uncover an entirely unexpected, novel function of Syt10 underlying the response of neurons to pathophysiologic activity and provide new therapeutic perspectives for neurological disorders.
Collapse
|
36
|
Antoniotti S, Ruffinatti FA, Torriano S, Luganini A, D'Alessandro R, Lovisolo D. REST levels affect the functional expression of voltage dependent calcium channels and the migratory activity in immortalized GnRH neurons. Neurosci Lett 2016; 629:19-25. [PMID: 27349310 DOI: 10.1016/j.neulet.2016.06.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/23/2016] [Indexed: 12/27/2022]
Abstract
The repressor element-1 silencing transcription factor (REST) has emerged as a key controller of neuronal differentiation and has been shown to play a critical role in the expression of the neuronal phenotype; however, much has still to be learned about its role at specific developmental stages and about the functional targets affected. Among these targets, calcium signaling mechanisms are critically dependent on the developmental stage and their full expression is a hallmark of the mature, functional neuron. We have analyzed the role played by REST in GN11 cells, an immortalized cell line derived from gonadotropin hormone releasing hormone (GnRH) neurons at an early developmental stage, electrically non-excitable and with a strong migratory activity. We show for the first time that functional voltage-dependent calcium channels are expressed in wild type GN11 cells; down-regulation of REST by a silencing approach shifts these cells towards a more differentiated phenotype, increasing the functional expression of P/Q-type channels and reducing their migratory potential.
Collapse
Affiliation(s)
- Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 10123, Torino, Italy; NIS Centre for Nanostructured Interfaces and Surfaces, University of Torino, Italy
| | | | - Simona Torriano
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 10123, Torino, Italy
| | - Anna Luganini
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 10123, Torino, Italy
| | - Rosalba D'Alessandro
- National Institute for Digestive Diseases De Bellis, via Turi 27, 70013, Castellana Grotte, Bari, Italy
| | - Davide Lovisolo
- Department of Life Sciences and Systems Biology, University of Torino, via Accademia Albertina 13, 10123, Torino, Italy; NIS Centre for Nanostructured Interfaces and Surfaces, University of Torino, Italy; Neuroscience Institute of Torino, Italy.
| |
Collapse
|
37
|
Pabst M, Braganza O, Dannenberg H, Hu W, Pothmann L, Rosen J, Mody I, van Loo K, Deisseroth K, Becker AJ, Schoch S, Beck H. Astrocyte Intermediaries of Septal Cholinergic Modulation in the Hippocampus. Neuron 2016; 90:853-65. [PMID: 27161528 DOI: 10.1016/j.neuron.2016.04.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 07/09/2015] [Accepted: 03/14/2016] [Indexed: 01/17/2023]
Abstract
The neurotransmitter acetylcholine, derived from the medial septum/diagonal band of Broca complex, has been accorded an important role in hippocampal learning and memory processes. However, the precise mechanisms whereby acetylcholine released from septohippocampal cholinergic neurons acts to modulate hippocampal microcircuits remain unknown. Here, we show that acetylcholine release from cholinergic septohippocampal projections causes a long-lasting GABAergic inhibition of hippocampal dentate granule cells in vivo and in vitro. This inhibition is caused by cholinergic activation of hilar astrocytes, which provide glutamatergic excitation of hilar inhibitory interneurons. These results demonstrate that acetylcholine release can cause slow inhibition of principal neuronal activity via astrocyte intermediaries.
Collapse
Affiliation(s)
- Milan Pabst
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Oliver Braganza
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Holger Dannenberg
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Wen Hu
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Leonie Pothmann
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Jurij Rosen
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Istvan Mody
- Department of Neurology, UCLA School of Medicine, 635 Charles Young Drive South, Los Angeles, CA 90095, USA
| | - Karen van Loo
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University and Howard Hughes Medical Institute, 318 Campus Drive, Stanford, CA 94305, USA
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Heinz Beck
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 25, 53105 Bonn, Germany.
| |
Collapse
|
38
|
|
39
|
Ondacova K, Karmazinova M, Lazniewska J, Weiss N, Lacinova L. Modulation of Cav3.2 T-type calcium channel permeability by asparagine-linked glycosylation. Channels (Austin) 2016; 10:175-84. [PMID: 26745591 DOI: 10.1080/19336950.2016.1138189] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Low-voltage-gated T-type calcium channels are expressed throughout the nervous system where they play an essential role in shaping neuronal excitability. Defects in T-type channel expression have been linked to various neuronal disorders including neuropathic pain and epilepsy. Currently, little is known about the cellular mechanisms controlling the expression and function of T-type channels. Asparagine-linked glycosylation has recently emerged as an essential signaling pathway by which the cellular environment can control expression of T-type channels. However, the role of N-glycans in the conducting function of T-type channels remains elusive. In the present study, we used human Cav3.2 glycosylation-deficient channels to assess the role of N-glycosylation on the gating of the channel. Patch-clamp recordings of gating currents revealed that N-glycans attached to hCav3.2 channels have a minimal effect on the functioning of the channel voltage-sensor. In contrast, N-glycosylation on specific asparagine residues may have an essential role in the conducting function of the channel by enhancing the channel permeability and / or the pore opening of the channel. Our data suggest that modulation of N-linked glycosylation of hCav3.2 channels may play an important physiological role, and could also support the alteration of T-type currents observed in disease states.
Collapse
Affiliation(s)
- Katarina Ondacova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| | - Maria Karmazinova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| | - Joanna Lazniewska
- b Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. , Prague , Czech Republic
| | - Norbert Weiss
- b Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. , Prague , Czech Republic
| | - Lubica Lacinova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| |
Collapse
|
40
|
LRP12 silencing during brain development results in cortical dyslamination and seizure sensitization. Neurobiol Dis 2015; 86:170-6. [PMID: 26639854 DOI: 10.1016/j.nbd.2015.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 10/20/2015] [Accepted: 11/26/2015] [Indexed: 02/04/2023] Open
Abstract
Correct positioning and differentiation of neurons during brain development is a key precondition for proper function. Focal cortical dysplasias (FCDs) are increasingly recognized as causes of therapy refractory epilepsies. Neuropathological analyses of respective surgical specimens from neurosurgery for seizure control often reveal aberrant cortical architecture and/or aberrantly shaped neurons in FCDs. However, the molecular pathogenesis particularly of FCDs with aberrant lamination (so-called FCD type I) is largely unresolved. Lipoproteins and particularly low-density lipoprotein receptor-related protein 12 (LRP12) are involved in brain development. Here, we have examined a potential role of LRP12 in the pathogenesis of FCDs. In vitro knockdown of LRP12 in primary neurons results in impaired neuronal arborization. In vivo ablation of LRP12 by intraventricularly in utero electroporated shRNAs elicits cortical maldevelopment, i.e. aberrant lamination by malpositioning of upper cortical layer neurons. Subsequent epilepsy phenotyping revealed pentylenetetrazol (PTZ)-induced seizures to be aggravated in cortical LRP12-silenced mice. Our data demonstrates IUE mediated cortical gene silencing as an excellent approach to study the role of distinct molecules for epilepsy associated focal brain lesions and suggests LRP12 and lipoprotein homeostasis as potential molecular target structures for the emergence of epilepsy-associated FCDs.
Collapse
|
41
|
van Loo KMJ, Schaub C, Pitsch J, Kulbida R, Opitz T, Ekstein D, Dalal A, Urbach H, Beck H, Yaari Y, Schoch S, Becker AJ. Zinc regulates a key transcriptional pathway for epileptogenesis via metal-regulatory transcription factor 1. Nat Commun 2015; 6:8688. [PMID: 26498180 PMCID: PMC4846312 DOI: 10.1038/ncomms9688] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 09/18/2015] [Indexed: 11/29/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common focal seizure disorder in adults. In many patients, transient brain insults, including status epilepticus (SE), are followed by a latent period of epileptogenesis, preceding the emergence of clinical seizures. In experimental animals, transcriptional upregulation of CaV3.2 T-type Ca(2+)-channels, resulting in an increased propensity for burst discharges of hippocampal neurons, is an important trigger for epileptogenesis. Here we provide evidence that the metal-regulatory transcription factor 1 (MTF1) mediates the increase of CaV3.2 mRNA and intrinsic excitability consequent to a rise in intracellular Zn(2+) that is associated with SE. Adeno-associated viral (rAAV) transfer of MTF1 into murine hippocampi leads to increased CaV3.2 mRNA. Conversely, rAAV-mediated expression of a dominant-negative MTF1 abolishes SE-induced CaV3.2 mRNA upregulation and attenuates epileptogenesis. Finally, data from resected human hippocampi surgically treated for pharmacoresistant TLE support the Zn(2+)-MTF1-CaV3.2 cascade, thus providing new vistas for preventing and treating TLE.
Collapse
Affiliation(s)
- Karen M. J. van Loo
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Christina Schaub
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Bonn 53105, Germany
- Department of Neurology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Julika Pitsch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Rebecca Kulbida
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Thoralf Opitz
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Dana Ekstein
- Department of Medical Neurobiology, IMRIC, Hebrew University–Hadassah School of Medicine, Jerusalem 91120, Israel
- Department of Neurology, Hadassah—Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Adam Dalal
- Department of Medical Neurobiology, IMRIC, Hebrew University–Hadassah School of Medicine, Jerusalem 91120, Israel
| | - Horst Urbach
- Department of Neuroradiology, Medical Center University of Freiburg, Freiburg 79106, Germany
| | - Heinz Beck
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Yoel Yaari
- Department of Medical Neurobiology, IMRIC, Hebrew University–Hadassah School of Medicine, Jerusalem 91120, Israel
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Albert J. Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| |
Collapse
|
42
|
Functional upregulation of the H2S/Cav3.2 channel pathway accelerates secretory function in neuroendocrine-differentiated human prostate cancer cells. Biochem Pharmacol 2015; 97:300-9. [DOI: 10.1016/j.bcp.2015.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022]
|
43
|
Synergy of direct and indirect cholinergic septo-hippocampal pathways coordinates firing in hippocampal networks. J Neurosci 2015; 35:8394-410. [PMID: 26041909 DOI: 10.1523/jneurosci.4460-14.2015] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The medial septum/diagonal band of Broca complex (MSDB) is a key structure that modulates hippocampal rhythmogenesis. Cholinergic neurons of the MSDB play a central role in generating and pacing theta-band oscillations in the hippocampal formation during exploration, novelty detection, and memory encoding. How precisely cholinergic neurons affect hippocampal network dynamics in vivo, however, has remained elusive. In this study, we show that stimulation of cholinergic MSDB neurons in urethane-anesthetized mice acts on hippocampal networks via two distinct pathways. A direct septo-hippocampal cholinergic projection causes increased firing of hippocampal inhibitory interneurons with concomitantly decreased firing of principal cells. In addition, cholinergic neurons recruit noncholinergic neurons within the MSDB. This indirect pathway is required for hippocampal theta synchronization. Activation of both pathways causes a reduction in pyramidal neuron firing and a more precise coupling to the theta oscillatory phase. These two anatomically and functionally distinct pathways are likely relevant for cholinergic control of encoding versus retrieval modes in the hippocampus.
Collapse
|
44
|
The Transcription Repressor REST in Adult Neurons: Physiology, Pathology, and Diseases. eNeuro 2015; 2:eN-REV-0010-15. [PMID: 26465007 PMCID: PMC4596026 DOI: 10.1523/eneuro.0010-15.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/11/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
REST [RE1-silencing transcription factor (also called neuron-restrictive silencer factor)] is known to repress thousands of possible target genes, many of which are neuron specific. To date, REST repression has been investigated mostly in stem cells and differentiating neurons. Current evidence demonstrates its importance in adult neurons as well. Low levels of REST, which are acquired during differentiation, govern the expression of specific neuronal phenotypes. REST-dependent genes encode important targets, including transcription factors, transmitter release proteins, voltage-dependent and receptor channels, and signaling proteins. Additional neuronal properties depend on miRNAs expressed reciprocally to REST and on specific splicing factors. In adult neurons, REST levels are not always low. Increases occur during aging in healthy humans. Moreover, extensive evidence demonstrates that prolonged stimulation with various agents induces REST increases, which are associated with the repression of neuron-specific genes with appropriate, intermediate REST binding affinity. Whether neuronal increases in REST are protective or detrimental remains a subject of debate. Examples of CA1 hippocampal neuron protection upon depolarization, and of neurodegeneration upon glutamate treatment and hypoxia have been reported. REST participation in psychiatric and neurological diseases has been shown, especially in Alzheimer’s disease and Huntington’s disease, as well as epilepsy. Distinct, complex roles of the repressor in these different diseases have emerged. In conclusion, REST is certainly very important in a large number of conditions. We suggest that the conflicting results reported for the role of REST in physiology, pathology, and disease depend on its complex, direct, and indirect actions on many gene targets and on the diverse approaches used during the investigations.
Collapse
|
45
|
Function and developmental origin of a mesocortical inhibitory circuit. Nat Neurosci 2015; 18:872-82. [DOI: 10.1038/nn.4020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023]
|
46
|
Terada Y, Fujimura M, Nishimura S, Tsubota M, Sekiguchi F, Kawabata A. Roles of Cav3.2 and TRPA1 channels targeted by hydrogen sulfide in pancreatic nociceptive processing in mice with or without acute pancreatitis. J Neurosci Res 2014; 93:361-9. [PMID: 25267397 DOI: 10.1002/jnr.23490] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/05/2014] [Accepted: 09/07/2014] [Indexed: 12/14/2022]
Abstract
Hydrogen sulfide (H(2)S), formed by multiple enzymes, including cystathionine-γ-lyase (CSE), targets Ca(v)3.2 T-type Ca(2+) channels (T channels) and transient receptor potential ankyrin-1 (TRPA1), facilitating somatic pain. Pancreatitis-related pain also appears to involve activation of T channels by H(2)S formed by the upregulated CSE. Therefore, this study investigates the roles of the Ca(v)3.2 isoform and/or TRPA1 in pancreatic nociception in the absence and presence of pancreatitis. In anesthetized mice, AP18, a TRPA1 inhibitor, abolished the Fos expression in the spinal dorsal horn caused by injection of a TRPA1 agonist into the pancreatic duct. As did mibefradil, a T-channel inhibitor, in our previous report, AP18 prevented the Fos expression following ductal NaHS, an H(2)S donor. In the mice with cerulein-induced acute pancreatitis, the referred hyperalgesia was suppressed by NNC 55-0396 (NNC), a selective T-channel inhibitor; zinc chloride; or ascorbic acid, known to inhibit Ca(v)3.2 selectively among three T-channel isoforms; and knockdown of Ca(v)3.2. In contrast, AP18 and knockdown of TRPA1 had no significant effect on the cerulein-induced referred hyperalgesia, although they significantly potentiated the antihyperalgesic effect of NNC at a subeffective dose. TRPA1 but not Ca(v)3.2 in the dorsal root ganglia was downregulated at a protein level in mice with cerulein-induced pancreatitis. The data indicate that TRPA1 and Ca(v)3.2 mediate the exogenous H(2)S-induced pancreatic nociception in naïve mice and suggest that, in the mice with pancreatitis, Ca(v)3.2 targeted by H(2)S primarily participates in the pancreatic pain, whereas TRPA1 is downregulated and plays a secondary role in pancreatic nociceptive signaling.
Collapse
Affiliation(s)
- Yuka Terada
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka, Japan
| | | | | | | | | | | |
Collapse
|
47
|
McClelland S, Brennan GP, Dubé C, Rajpara S, Iyer S, Richichi C, Bernard C, Baram TZ. The transcription factor NRSF contributes to epileptogenesis by selective repression of a subset of target genes. eLife 2014; 3:e01267. [PMID: 25117540 PMCID: PMC4129437 DOI: 10.7554/elife.01267] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The mechanisms generating epileptic neuronal networks following insults such as severe seizures are unknown. We have previously shown that interfering with the function of the neuron-restrictive silencer factor (NRSF/REST), an important transcription factor that influences neuronal phenotype, attenuated development of this disorder. In this study, we found that epilepsy-provoking seizures increased the low NRSF levels in mature hippocampus several fold yet surprisingly, provoked repression of only a subset (∼10%) of potential NRSF target genes. Accordingly, the repressed gene-set was rescued when NRSF binding to chromatin was blocked. Unexpectedly, genes selectively repressed by NRSF had mid-range binding frequencies to the repressor, a property that rendered them sensitive to moderate fluctuations of NRSF levels. Genes selectively regulated by NRSF during epileptogenesis coded for ion channels, receptors, and other crucial contributors to neuronal function. Thus, dynamic, selective regulation of NRSF target genes may play a role in influencing neuronal properties in pathological and physiological contexts. DOI:http://dx.doi.org/10.7554/eLife.01267.001 Epilepsy is a common brain disease that can cause disabling seizures. During a seizure, brain cells send out abnormal signals, which can mean that people having seizures may be unaware of their surroundings and may fall or otherwise injure themselves. Individuals with epilepsy develop changes in their brain cells and in the circuits that connect these cells together. Some people develop epilepsy because they have mutations in genes. Others develop the condition after an injury or a long seizure, which leads to changes in gene expression and therefore changes to the brain's cells and circuits. In 2011, researchers found that a protein that normally switches off the expression of certain genes during brain development, but which is almost absent in the adult brain, may run amok after a seizure. The level of this protein—a transcription factor called NRSF—increased in the brains of rats that had been caused to have a seizure. A long provoked seizure caused many of the rats to develop epilepsy. But, if NRSF was blocked after the original seizure, the rats were less likely to have further seizures later on. Now McClelland et al., including several of the researchers involved in the 2011 work, have examined what normally happens to the expression of genes after a seizure and what happens when the NRSF transcription factor is blocked. McClelland et al. found that only a small subset—about 10%—of the genes that can theoretically be silenced by NRSF are switched off in the brain when this protein's levels increase after a seizure. The increased NRSF levels, unexpectedly, did not affect the genes that bind tightly to this transcription factor. Nor did NRSF affect genes that bind loosely. Instead, the genes that the transcription factor binds to with an intermediate strength were the ones that were switched off. McClelland et al. suggest that this ‘mid-range binding’ to NRSF allows the expression of these genes to be increased or decreased in response to there being more or less NRSF in the cell. Genes that bind tightly to NRSF are likely to already have a lot of NRSF bound and are therefore already switched off; and loosely-binding genes would likely need even more NRSF before they are switched off. The subset of genes that were switched off by the increased levels of NRSF after a seizure code for a number of proteins that brain cells need to be able to effectively send and receive messages. Blocking the ability of NRSF to bind to these genes and switch them off may help to prevent the brain changes that cause epilepsy. DOI:http://dx.doi.org/10.7554/eLife.01267.002
Collapse
Affiliation(s)
- Shawn McClelland
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Gary P Brennan
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Celine Dubé
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Seeta Rajpara
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Shruti Iyer
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Cristina Richichi
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| | - Christophe Bernard
- Laboratoire Epilepsie et Cognition, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, United States Department of Pediatrics, University of California, Irvine, Irvine, United States Department of Neurology, University of California, Irvine, Irvine, United States
| |
Collapse
|
48
|
Sellak H, Zhou C, Liu B, Chen H, Lincoln TM, Wu S. Transcriptional regulation of α1H T-type calcium channel under hypoxia. Am J Physiol Cell Physiol 2014; 307:C648-56. [PMID: 25099734 DOI: 10.1152/ajpcell.00210.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The low-voltage-activated T-type Ca(2+) channels play an important role in mediating the cellular responses to altered oxygen tension. Among three T-type channel isoforms, α1G, α1H, and α1I, only α1H was found to be upregulated under hypoxia. However, mechanisms underlying such hypoxia-dependent isoform-specific gene regulation remain incompletely understood. We, therefore, studied the hypoxia-dependent transcriptional regulation of α1G and α1H gene promoters with the aim to identify the functional hypoxia-response elements (HREs). In rat pulmonary artery smooth muscle cells (PASMCs) and pheochromocytoma (PC12) cells after hypoxia (3% O2) exposure, we observed a prominent increase in α1H mRNA at 12 h along with a significant rise in α1H-mediated T-type current at 24 and 48 h. We then cloned two promoter fragments from the 5'-flanking regions of rat α1G and α1H gene, 2,000 and 3,076 bp, respectively, and inserted these fragments into a luciferase reporter vector. Transient transfection of PASMCs and PC12 cells with these recombinant constructs and subsequent luciferase assay revealed a significant increase in luciferase activity from the reporter containing the α1H, but not α1G, promoter fragment under hypoxia. Using serial deletion and point mutation analysis strategies, we identified a functional HRE at site -1,173cacgc-1,169 within the α1H promoter region. Furthermore, an electrophoretic mobility shift assay using this site as a DNA probe demonstrated an increased binding activity to nuclear protein extracts from the cells after hypoxia exposure. Taken together, these findings indicate that hypoxia-induced α1H upregulation involves binding of hypoxia-inducible factor to an HRE within the α1H promoter region.
Collapse
Affiliation(s)
- Hassan Sellak
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia
| | - Chun Zhou
- Center for Lung Biology, University of South Alabama, Mobile, Alabama; Department of Pharmacology, University of South Alabama, Mobile, Alabama; and
| | - Bainan Liu
- Center for Lung Biology, University of South Alabama, Mobile, Alabama; Department of Pharmacology, University of South Alabama, Mobile, Alabama; and
| | - Hairu Chen
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia
| | - Thomas M Lincoln
- Department of Physiology, University of South Alabama, Mobile, Alabama
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia;
| |
Collapse
|
49
|
Rossignol E, Kobow K, Simonato M, Loeb JA, Grisar T, Gilby KL, Vinet J, Kadam SD, Becker AJ. WONOEP appraisal: new genetic approaches to study epilepsy. Epilepsia 2014; 55:1170-86. [PMID: 24965021 PMCID: PMC4126888 DOI: 10.1111/epi.12692] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2014] [Indexed: 12/19/2022]
Abstract
New genetic investigation techniques, including next-generation sequencing, epigenetic profiling, cell lineage mapping, targeted genetic manipulation of specific neuronal cell types, stem cell reprogramming, and optogenetic manipulations within epileptic networks are progressively unraveling the mysteries of epileptogenesis and ictogenesis. These techniques have opened new avenues to discover the molecular basis of epileptogenesis and to study the physiologic effects of mutations in epilepsy-associated genes on a multilayer level, from cells to circuits. This manuscript reviews recently published applications of these new genetic technologies in the study of epilepsy, as well as work presented by the authors at the genetic session of the XII Workshop on the Neurobiology of Epilepsy (WONOEP 2013) in Quebec, Canada. Next-generation sequencing is providing investigators with an unbiased means to assess the molecular causes of sporadic forms of epilepsy and has revealed the complexity and genetic heterogeneity of sporadic epilepsy disorders. To assess the functional impact of mutations in these newly identified genes on specific neuronal cell types during brain development, new modeling strategies in animals, including conditional genetics in mice and in utero knock-down approaches, are enabling functional validation with exquisite cell-type and temporal specificity. In addition, optogenetics, using cell-type-specific Cre recombinase driver lines, is enabling investigators to dissect networks involved in epilepsy. In addition, genetically encoded cell-type labeling is providing new means to assess the role of the nonneuronal components of epileptic networks such as glial cells. Furthermore, beyond its role in revealing coding variants involved in epileptogenesis, next-generation sequencing can be used to assess the epigenetic modifications that lead to sustained network hyperexcitability in epilepsy, including methylation changes in gene promoters and noncoding ribonucleic acid (RNA) involved in modifying gene expression following seizures. In addition, genetically based bioluminescent reporters are providing new opportunities to assess neuronal activity and neurotransmitter levels both in vitro and in vivo in the context of epilepsy. Finally, genetically rederived neurons generated from patient induced pluripotent stem cells and genetically modified zebrafish have become high-throughput means to investigate disease mechanisms and potential new therapies. Genetics has changed the field of epilepsy research considerably, and is paving the way for better diagnosis and therapies for patients with epilepsy.
Collapse
Affiliation(s)
- Elsa Rossignol
- Pediatric & Neuroscience Dept. & Brain Disease Research Group, CHU Ste-Justine, Montreal, Canada
| | - Katja Kobow
- Dept. of Neuropathology, Univ. Hospital Erlangen, Germany
| | - Michele Simonato
- Dept. of Medical Sciences (Pharmacology), Univ. of Ferrara, Italy
| | - Jeffrey A. Loeb
- Dept. of Neurology & Rehabilitation, Univ. of Illinois, Chicago, USA
| | | | - Krista L. Gilby
- Dept. of Medicine, Royal Hospital, The Melbourne Brain Centre, Univ. of Melbourne, Australia
| | - Jonathan Vinet
- Dept. of Neural, Biomedical, Metabolic & Neural Sciences, Univ. of Modena, Italy
| | - Shilpa D. Kadam
- Depts. of Neuroscience and Neurology, Kennedy Krieger & Johns Hopkins Univ. School of Medicine of Baltimore, USA
| | | |
Collapse
|
50
|
Kulbida R, Wang Y, Mandelkow EM, Schoch S, Becker AJ, van Loo KMJ. Molecular imaging reveals epileptogenic Ca2+-channel promoter activation in hippocampi of living mice. Brain Struct Funct 2014; 220:3067-73. [PMID: 24889163 DOI: 10.1007/s00429-014-0801-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 05/14/2014] [Indexed: 10/25/2022]
Abstract
Focal epilepsies often originate in the hippocampal formation of the temporal lobe (temporal lobe epilepsy) and are generally acquired after transient brain insults. Such insults induce cellular and structural reorganization processes of the hippocampus, referred to as epileptogenesis that finally convert the brain spontaneous epileptic. Here, we developed a new molecular imaging strategy in a state-of-the-art animal model to provide insights into key epileptogenic mechanisms. Our new approach combines recombinant adeno-associated virus (rAAV) gene delivery with in vivo bioluminescence imaging. rAAV particles harboring the luciferase reporter gene under control of the minimal T type Ca(2+)-channel subunit Ca V 3.2-promoter were generated and injected stereotaxically in the hippocampal region of mice. Bioluminescent signals, corresponding to Ca V 3.2 promoter activation, were imaged in vivo in the pilocarpine model of status epilepticus (SE). We detected activation of key Ca V 3.2 promoter motifs at 3 and 10 days after SE but not after the onset of chronic seizures. These data suggest Ca V 3.2 promoter activation as novel anti-epileptogenic target. In more general terms, we have established an experimental approach that allows to follow cerebral gene promoter dynamics longitudinally and to correlate this activity to behavioral parameters in the same mice.
Collapse
Affiliation(s)
- Rebecca Kulbida
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53105, Bonn, Germany
| | | | | | | | | | | |
Collapse
|