1
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of intracellular FGF14 (iFGF14) increases excitability of mature hippocampal pyramidal neurons. J Gen Physiol 2025; 157:e202413597. [PMID: 40323232 PMCID: PMC12051480 DOI: 10.1085/jgp.202413597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 01/30/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Mutations in FGF14, which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with deficits in motor coordination and cognitive function. Mice lacking iFGF14 (Fgf14-/-) display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to a hyperpolarizing shift in the voltage-dependence of voltage-gated Na+ (Nav) current steady-state inactivation. Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal pyramidal neurons. Current-clamp recordings from CA1 pyramidal neurons in acute in vitro slices, however, revealed that evoked repetitive firing rates were higher in Fgf14-/- than in wild type (WT) cells. Also, in contrast with Purkinje neurons, voltage-clamp recordings demonstrated that the loss of iFGF14 did not affect the voltage dependence of steady-state inactivation of the Nav currents in CA1 pyramidal neurons. In addition, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, immunohistochemical experiments revealed that loss of iFGF14 does not disrupt the localization or alter the normalized distribution of α-Nav1.6 or α-ankyrin G labeling along the axon initial segments (AIS) of mature hippocampal CA1 neurons in situ. However, the integrated intensities of α-Nav1.6 labeling were significantly higher along the AIS of Fgf14-/-, compared with WT, adult hippocampal CA1 pyramidal neurons, consistent with the marked increase in the excitability of CA1 neurons with the loss of iFGF14.
Collapse
Affiliation(s)
- Joseph L. Ransdell
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yarimar Carrasquillo
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marie K. Bosch
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca L. Mellor
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Ornitz
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Köster PA, Leipold E, Tigerholm J, Maxion A, Namer B, Stiehl T, Lampert A. Nociceptor sodium channels shape subthreshold phase, upstroke, and shoulder of action potentials. J Gen Physiol 2025; 157:e202313526. [PMID: 39836077 PMCID: PMC11748974 DOI: 10.1085/jgp.202313526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Voltage-gated sodium channels (VGSCs) in the peripheral nervous system shape action potentials (APs) and thereby support the detection of sensory stimuli. Most of the nine mammalian VGSC subtypes are expressed in nociceptors, but predominantly, three are linked to several human pain syndromes: while Nav1.7 is suggested to be a (sub-)threshold channel, Nav1.8 is thought to support the fast AP upstroke. Nav1.9, as it produces large persistent currents, is attributed a role in determining the resting membrane potential. We characterized the gating of Nav1.1-Nav1.3 and Nav1.5-Nav1.9 in manual patch clamp with a focus on the AP subthreshold depolarization phase. Nav1.9 exhibited the most hyperpolarized activation, while its fast inactivation resembled the depolarized inactivation of Nav1.8. For some VGSCs (e.g., Nav1.1 and Nav1.2), a positive correlation between ramp current and window current was detected. Using a modified Hodgkin-Huxley model that accounts for the time needed for inactivation to occur, we used the acquired data to simulate two nociceptive nerve fiber types (an Aδ- and a mechano-insensitive C-nociceptor) containing VGSC conductances according to published human RNAseq data. Our simulations suggest that Nav1.9 is supporting both the AP upstroke and its shoulder. A reduced threshold for AP generation was induced by enhancing Nav1.7 conductivity or shifting its activation to more hyperpolarized potentials, as observed in Nav1.7-related pain disorders. Here, we provide a comprehensive, comparative functional characterization of VGSCs relevant in nociception and describe their gating with Hodgkin-Huxley-like models, which can serve as a tool to study their specific contributions to AP shape and sodium channel-related diseases.
Collapse
Affiliation(s)
- Phil Alexander Köster
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care and CBBM-Center of Brain, Behavior and Metabolism, University of Luebeck, Lübeck, Germany
| | - Jenny Tigerholm
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
| | - Anna Maxion
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Barbara Namer
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Thomas Stiehl
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
- Institute for Computational Biomedicine and Disease Modelling With Focus on Phase Transitions Between Phenotypes, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| |
Collapse
|
3
|
Frazee A, Zybura A, Cummins TR. Differential Regulation of Nav1.1 and SCN1A Disease Mutant Sodium Current Properties by Fibroblast Growth Factor Homologous Factors. Cells 2025; 14:291. [PMID: 39996763 PMCID: PMC11853998 DOI: 10.3390/cells14040291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Fibroblast growth factor homologous factors (FHFs) regulate the activity of several different voltage-gated sodium channels (Navs). However, more work is needed to determine how specific FHF isoforms and variants affect the properties of different Nav isoforms. In addition, it is not known if FHFs can differentially modulate the properties of Nav variants associated with disease. Here, we investigated the effects of FHF2A and FHF2B on Nav1.1 properties as well as on a familial hemiplegic migraine 3 (FHM3) causing mutation in this channel, F1774S. We found that FHF2A, but not 2B, induced prominent long-term inactivation (LTI) in the wild-type (WT) Nav1.1. Interestingly, FHF2A induced LTI in the F1774S FHM3 mutant channel to a greater extent than in the WT. Furthermore, persistent currents caused by the F1774S mutation were attenuated by the co-expression of FHF2A, leading to a possible rescue of the mutant channel phenotype. By contrast, the P1894L mutation, which is associated with epilepsy and mild intellectual disability, greatly attenuated the LTI induced by FHF2A. Overall, our data show for the first time that FHF2A might be a significant modulator of Nav1.1 that can differentially modulate the impact of Nav1.1 disease-associated mutations.
Collapse
Affiliation(s)
- Ashley Frazee
- Biology Department, School of Science, Indiana University Indianapolis, Indianapolis, IN 46202, USA; (A.F.)
| | - Agnes Zybura
- Biology Department, School of Science, Indiana University Indianapolis, Indianapolis, IN 46202, USA; (A.F.)
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Theodore R. Cummins
- Biology Department, School of Science, Indiana University Indianapolis, Indianapolis, IN 46202, USA; (A.F.)
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Dvorak NM, Wadsworth PA, Aquino-Miranda G, Wang P, Engelke DS, Zhou J, Nguyen N, Singh AK, Aceto G, Haghighijoo Z, Smith II, Goode N, Zhou M, Avchalumov Y, Troendle EP, Tapia CM, Chen H, Powell RT, Baumgartner TJ, Singh J, Koff L, Di Re J, Wadsworth AE, Marosi M, Azar MR, Elias K, Lehmann P, Mármol Contreras YM, Shah P, Gutierrez H, Green TA, Ulmschneider MB, D'Ascenzo M, Stephan C, Cui G, Do Monte FH, Zhou J, Laezza F. Enhanced motivated behavior mediated by pharmacological targeting of the FGF14/Na v1.6 complex in nucleus accumbens neurons. Nat Commun 2025; 16:110. [PMID: 39747162 PMCID: PMC11696184 DOI: 10.1038/s41467-024-55554-7] [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: 03/30/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Protein/protein interactions (PPI) play crucial roles in neuronal functions. Yet, their potential as drug targets for brain disorders remains underexplored. The fibroblast growth factor 14 (FGF14)/voltage-gated Na+ channel 1.6 (Nav1.6) complex regulates excitability of medium spiny neurons (MSN) of the nucleus accumbens (NAc), a central hub of reward circuitry that controls motivated behaviors. Here, we identified compound 1028 (IUPAC: ethyl 3-(2-(3-(hydroxymethyl)-1H-indol-1-yl)acetamido)benzoate), a brain-permeable small molecule that targets FGF14R117, a critical residue located within a druggable pocket at the FGF14/Nav1.6 PPI interface. We found that 1028 modulates FGF14/Nav1.6 complex assembly and depolarizes the voltage-dependence of Nav1.6 channel inactivation with nanomolar potency by modulating the intramolecular interaction between the III-IV linker and C-terminal domain of the Nav1.6 channel. Consistent with the compound's effects on Nav1.6 channel inactivation, 1028 enhances MSN excitability ex vivo and accumbal neuron firing rate in vivo in murine models. Systemic administration of 1028 maintains behavioral motivation preferentially during motivationally deficient conditions in murine models. These behavioral effects were abrogated by in vivo gene silencing of Fgf14 in the NAc and were accompanied by a selective reduction in accumbal dopamine levels during reward consumption in murine models. These findings underscore the potential to selectively regulate complex behaviors associated with neuropsychiatric disorders through targeting of PPIs in neurons.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Paul A Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, Stanford Medicine, Stanford, CA, USA
| | - Guillermo Aquino-Miranda
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Douglas S Engelke
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Jingheng Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Nghi Nguyen
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Zahra Haghighijoo
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Isabella I Smith
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Nana Goode
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mingxiang Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yosef Avchalumov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Evan P Troendle
- Department of Chemistry, King's College London 7 Trinity Street, London, UK
| | - Cynthia M Tapia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Haiying Chen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Reid T Powell
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Timothy J Baumgartner
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jully Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Leandra Koff
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jessica Di Re
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ann E Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mate Marosi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Marc R Azar
- Behavioral Pharma Inc., 505 Coast Blvd. South, Suite 212, La Jolla, CA, USA
| | - Kristina Elias
- Behavioral Pharma Inc., 505 Coast Blvd. South, Suite 212, La Jolla, CA, USA
| | - Paul Lehmann
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Poonam Shah
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hector Gutierrez
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas A Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Clifford Stephan
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Fabricio H Do Monte
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
5
|
Rivas LJ, Uribe RA. Fibroblast Growth Factor (FGF) 13. Differentiation 2024; 140:100814. [PMID: 39332965 PMCID: PMC12011324 DOI: 10.1016/j.diff.2024.100814] [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: 11/21/2023] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Fibroblast Growth Factor (FGF) 13, also referred to as FGF homologous factor (FHF) 2, is a member of the FGF11 subfamily that is characterized as having sequence similarities to classical FGF receptor (FGFR)-binding FGFs, but functionally do not bind FGFRs. In this primer mini-review, we summarize current knowledge regarding FGF13 expression, mutant analyses, and gene and protein structure. Similar to other FHFs, FGF13 has been considered a non-secreted protein that lacks an amino signal and is prominently expressed in developing and mature neurons of the central and peripheral nervous systems, as well as the heart. The expression of FGF13 is not limited to early embryonic stages and has been shown to persist in adult tissues. As well, FGF13 is known to localize subcellularly, both within the cytoplasm and the nucleus. FGF13 is extremely adaptable, as it interacts with MAPK scaffolding protein islet brain 2 (IB2), stabilizes microtubules, or binds to voltage-gated sodium channels. Fgf13 mutant mouse lines display various neurological pathologies. Through sequence mapping, FGF13 is considered a candidate causative gene that is mutated in multiple human X-linked neurological diseases.
Collapse
Affiliation(s)
- Lucia J Rivas
- Department of Biosciences, Rice University, Houston, TX, United States; Biochemistry and Cell Biology Graduate Program, Rice University, Houston, TX, United States
| | - Rosa A Uribe
- Department of Biosciences, Rice University, Houston, TX, United States; Biochemistry and Cell Biology Graduate Program, Rice University, Houston, TX, United States.
| |
Collapse
|
6
|
Ransdell JL, Brown SP, Xiao M, Ornitz DM, Nerbonne JM. In Vivo Expression of an SCA27A-linked FGF14 Mutation Results in Haploinsufficiency and Impaired Firing of Cerebellar Purkinje Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620253. [PMID: 39484407 PMCID: PMC11527103 DOI: 10.1101/2024.10.25.620253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Autosomal dominant mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), underlie spinocerebellar ataxia type 27A (SCA27A), a devastating multisystem disorder resulting in progressive deficits in motor coordination and cognitive function. Mice lacking iFGF14 ( Fgf14 -/- ) exhibit similar phenotypes, which have been linked to iFGF14-mediated modulation of the voltage-gated sodium (Nav) channels that control the high frequency repetitive firing of Purkinje neurons, the main output neurons of the cerebellar cortex. To investigate the pathophysiological mechanisms underlying SCA27A, we developed a targeted knock-in strategy to introduce the first point mutation identified in FGF14 into the mouse Fgf14 locus ( Fgf14 F145S ), we determined the impact of in vivo expression of the mutant Fgf14 F145S allele on the motor performance of adult animals and on the firing properties of mature Purkinje neurons in acute cerebellar slices. Electrophysiological experiments revealed that repetitive firing rates are attenuated in adult Fgf14 F145S/+ cerebellar Purkinje neurons, attributed to a hyperpolarizing shift in the voltage-dependence of steady-state inactivation of Nav channels. More severe effects on firing properties and Nav channel inactivation were observed in homozygous Fgf14 F145S/F145S Purkinje neurons. Interestingly, the electrophysiological phenotypes identified in adult Fgf14 F145S/+ and Fgf14 F145S/F145S cerebellar Purkinje neurons mirror those observed in heterozygous Fgf14 +/- and homozygous Fgf14 -/- Purkinje neurons, respectively, suggesting that the mutation results in the loss of the iFGF14 protein. Western blot analysis of lysates from adult heterozygous Fgf14 F145S/+ and homozygous Fgf14 F145S/F145S animals revealed reduced or undetectable, respectively, iFGF14 expression, supporting the hypothesis that the mutant allele results in loss of the iFGF14 protein and that haploinsufficiency underlies SCA27A neurological phenotypes.
Collapse
|
7
|
Goldfarb M. Fibroblast growth factor homologous factors: canonical and non-canonical mechanisms of action. J Physiol 2024; 602:4097-4110. [PMID: 39083261 DOI: 10.1113/jp286313] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Since their discovery nearly 30 years ago, fibroblast growth factor homologous factors (FHFs) are now known to control the functionality of excitable tissues through a range of mechanisms. Nervous and cardiac system dysfunctions are caused by loss- or gain-of-function mutations in FHF genes. The best understood 'canonical' targets for FHF action are voltage-gated sodium channels, and recent studies have expanded the repertoire of ways that FHFs modulate sodium channel gating. Additional 'non-canonical' functions of FHFs in excitable and non-excitable cells, including cancer cells, have been reported over the past dozen years. This review summarizes and evaluates reported canonical and non-canonical FHF functions.
Collapse
Affiliation(s)
- Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, New York, New York, USA
- Biology Program, The Graduate Center City University, New York, New York, USA
| |
Collapse
|
8
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of Intracellular Fibroblast Growth Factor 14 (iFGF14) Increases the Excitability of Mature Hippocampal and Cortical Pyramidal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592532. [PMID: 38746081 PMCID: PMC11092765 DOI: 10.1101/2024.05.04.592532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with progressive deficits in motor coordination and cognitive function. Mice ( Fgf14 -/- ) lacking iFGF14 display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to the loss of iFGF14-mediated regulation of the voltage-dependence of inactivation of the fast transient component of the voltage-gated Na + (Nav) current, I NaT . Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal and cortical pyramidal neurons. Current-clamp recordings from adult mouse hippocampal CA1 pyramidal neurons in acute in vitro slices, however, revealed that repetitive firing rates were higher in Fgf14 -/- , than in wild type (WT), cells. In addition, the waveforms of individual action potentials were altered in Fgf14 -/- hippocampal CA1 pyramidal neurons, and the loss of iFGF14 reduced the time delay between the initiation of axonal and somal action potentials. Voltage-clamp recordings revealed that the loss of iFGF14 altered the voltage-dependence of activation, but not inactivation, of I NaT in CA1 pyramidal neurons. Similar effects of the loss of iFGF14 on firing properties were evident in current-clamp recordings from layer 5 visual cortical pyramidal neurons. Additional experiments demonstrated that the loss of iFGF14 does not alter the distribution of anti-Nav1.6 or anti-ankyrin G immunofluorescence labeling intensity along the axon initial segments (AIS) of mature hippocampal CA1 or layer 5 visual cortical pyramidal neurons in situ . Taken together, the results demonstrate that, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, the loss of iFGF14 does not disrupt AIS architecture or Nav1.6 localization/distribution along the AIS of mature hippocampal (or cortical) pyramidal neurons in situ .
Collapse
|
9
|
Libé-Philippot B, Lejeune A, Wierda K, Louros N, Erkol E, Vlaeminck I, Beckers S, Gaspariunaite V, Bilheu A, Konstantoulea K, Nyitrai H, De Vleeschouwer M, Vennekens KM, Vidal N, Bird TW, Soto DC, Jaspers T, Dewilde M, Dennis MY, Rousseau F, Comoletti D, Schymkowitz J, Theys T, de Wit J, Vanderhaeghen P. LRRC37B is a human modifier of voltage-gated sodium channels and axon excitability in cortical neurons. Cell 2023; 186:5766-5783.e25. [PMID: 38134874 PMCID: PMC10754148 DOI: 10.1016/j.cell.2023.11.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) β-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Amélie Lejeune
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Nikolaos Louros
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Emir Erkol
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Ine Vlaeminck
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Sofie Beckers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Vaiva Gaspariunaite
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Angéline Bilheu
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Katerina Konstantoulea
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Hajnalka Nyitrai
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Matthias De Vleeschouwer
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Niels Vidal
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Thomas W Bird
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Daniela C Soto
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Tom Jaspers
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Maarten Dewilde
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Megan Y Dennis
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Tom Theys
- KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Research Group Experimental Neurosurgery and Neuroanatomy, KUL, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium.
| |
Collapse
|
10
|
Dvorak NM, Di Re J, Vasquez TES, Marosi M, Shah P, Contreras YMM, Bernabucci M, Singh AK, Stallone J, Green TA, Laezza F. Fibroblast growth factor 13-mediated regulation of medium spiny neuron excitability and cocaine self-administration. Front Neurosci 2023; 17:1294567. [PMID: 38099204 PMCID: PMC10720079 DOI: 10.3389/fnins.2023.1294567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023] Open
Abstract
Cocaine use disorder (CUD) is a prevalent neuropsychiatric disorder with few existing treatments. Thus, there is an unmet need for the identification of new pharmacological targets for CUD. Previous studies using environmental enrichment versus isolation paradigms have found that the latter induces increased cocaine self-administration with correlative increases in the excitability of medium spiny neurons (MSN) of the nucleus accumbens shell (NAcSh). Expanding upon these findings, we sought in the present investigation to elucidate molecular determinants of these phenomena. To that end, we first employed a secondary transcriptomic analysis and found that cocaine self-administration differentially regulates mRNA for fibroblast growth factor 13 (FGF13), which codes for a prominent auxiliary protein of the voltage-gated Na+ (Nav) channel, in the NAcSh of environmentally enriched rats (i.e., resilient behavioral phenotype) compared to environmentally isolated rats (susceptible phenotype). Based upon this finding, we used in vivo genetic silencing to study the causal functional and behavioral consequences of knocking down FGF13 in the NAcSh. Functional studies revealed that knockdown of FGF13 in the NAcSh augmented excitability of MSNs by increasing the activity of Nav channels. These electrophysiological changes were concomitant with a decrease in cocaine demand elasticity (i.e., susceptible phenotype). Taken together, these data support FGF13 as being protective against cocaine self-administration, which positions it well as a pharmacological target for CUD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Thomas A. Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
11
|
Abstract
Voltage-gated sodium channel Nav1.6 plays a crucial role in neuronal firing in the central nervous system (CNS). Aberrant function of Nav1.6 may lead to epilepsy and other neurological disorders. Specific inhibitors of Nav1.6 thus have therapeutic potentials. Here we present the cryo-EM structure of human Nav1.6 in the presence of auxiliary subunits β1 and fibroblast growth factor homologous factor 2B (FHF2B) at an overall resolution of 3.1 Å. The overall structure represents an inactivated state with closed pore domain (PD) and all "up" voltage-sensing domains. A conserved carbohydrate-aromatic interaction involving Trp302 and Asn326, together with the β1 subunit, stabilizes the extracellular loop in repeat I. Apart from regular lipids that are resolved in the EM map, an unprecedented Y-shaped density that belongs to an unidentified molecule binds to the PD, revealing a potential site for developing Nav1.6-specific blockers. Structural mapping of disease-related Nav1.6 mutations provides insights into their pathogenic mechanism.
Collapse
|
12
|
Altered integration of excitatory inputs onto the basal dendrites of layer 5 pyramidal neurons in a mouse model of Fragile X syndrome. Proc Natl Acad Sci U S A 2023; 120:e2208963120. [PMID: 36595706 PMCID: PMC9926222 DOI: 10.1073/pnas.2208963120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Layer 5 (L5) pyramidal neurons receive predictive and sensory inputs in a compartmentalized manner at their apical and basal dendrites, respectively. To uncover how integration of sensory inputs is affected in autism spectrum disorders (ASD), we used two-photon glutamate uncaging to activate spines in the basal dendrites of L5 pyramidal neurons from a mouse model of Fragile X syndrome (FXS), the most common genetic cause of ASD. While subthreshold excitatory inputs integrate linearly in wild-type animals, surprisingly those with FXS summate sublinearly, contradicting what would be expected of sensory hypersensitivity classically associated with ASD. We next investigated the mechanism underlying this sublinearity by performing knockdown of the regulatory β4 subunit of BK channels, which rescued the synaptic integration, a result that was corroborated with numerical simulations. Taken together, these findings suggest that there is a differential impairment in the integration of feedforward sensory and feedback predictive inputs in L5 pyramidal neurons in FXS and potentially other forms of ASD, as a result of specifically localized subcellular channelopathies. These results challenge the traditional view that FXS and other ASD are characterized by sensory hypersensitivity, proposing instead a hyposensitivity of sensory inputs and hypersensitivity of predictive inputs onto cortical neurons.
Collapse
|
13
|
Effraim PR, Estacion M, Zhao P, Sosniak D, Waxman SG, Dib-Hajj SD. Fibroblast growth factor homologous factor 2 attenuates excitability of DRG neurons. J Neurophysiol 2022; 128:1258-1266. [PMID: 36222860 PMCID: PMC9909838 DOI: 10.1152/jn.00361.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor homologous factors (FHFs) are cytosolic members of the superfamily of the FGF proteins. Four members of this subfamily (FHF1-4) are differentially expressed in multiple tissues in an isoform-dependent manner. Mutations in FHF proteins have been associated with multiple neurological disorders. FHF proteins bind to the COOH terminus of voltage-gated sodium (Nav) channels and regulate current amplitude and gating properties of these channels. FHF2, which is expressed in dorsal root ganglia (DRG) neurons, has two main splicing isoforms: FHF2A and FHF2B, which differ in the length and sequence of their NH2 termini, have been shown to differentially regulate gating properties of Nav1.7, a channel that is a major driver of DRG neuron firing. FHF2 expression levels are downregulated after peripheral nerve axotomy, which suggests that they may regulate neuronal excitability via an action on Nav channels after injury. We have previously shown that knockdown of FHF2 leads to gain-of-function changes in Nav1.7 gating properties: enhanced repriming, increased current density, and hyperpolarized activation. From this we posited that knockdown of FHF2 might also lead to DRG hyperexcitability. Here we show that knockdown of either FHF2A alone or all isoforms of FHF2 results in increased DRG neuron excitability. In addition, we demonstrate that supplementation of FHF2A and FHF2B reduces DRG neuron excitability. Overexpression of FHF2A or FHF2B also reduced excitability of DRG neurons treated with a cocktail of inflammatory mediators, a model of inflammatory pain. Our data suggest that increased neuronal excitability after nerve injury might be triggered, in part, via a loss of FHF2-Nav1.7 interaction.NEW & NOTEWORTHY FHF2 is known to bind to and modulate the function of Nav1.7. FHF2 expression is also reduced after nerve injury. We demonstrate that knockdown of FHF2 expression increases DRG neuronal excitability. More importantly, overexpression of FHF2 reduces DRG excitability in basal conditions and in the presence of inflammatory mediators (a model of inflammatory pain). These results suggest that FHF2 could potentially be used as a tool to reduce DRG neuronal excitability and to treat pain.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
14
|
Chakouri N, Rivas S, Roybal D, Yang L, Diaz J, Hsu A, Mahling R, Chen BX, Owoyemi JO, DiSilvestre D, Sirabella D, Corneo B, Tomaselli GF, Dick IE, Marx SO, Ben-Johny M. Fibroblast growth factor homologous factors serve as a molecular rheostat in tuning arrhythmogenic cardiac late sodium current. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1-13. [PMID: 35662881 PMCID: PMC9161660 DOI: 10.1038/s44161-022-00060-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/04/2022] [Indexed: 05/20/2023]
Abstract
Voltage-gated sodium (Nav1.5) channels support the genesis and brisk spatial propagation of action potentials in the heart. Disruption of NaV1.5 inactivation results in a small persistent Na influx known as late Na current (I Na,L), which has emerged as a common pathogenic mechanism in both congenital and acquired cardiac arrhythmogenic syndromes. Here, using low-noise multi-channel recordings in heterologous systems, LQTS3 patient-derived iPSCs cardiomyocytes, and mouse ventricular myocytes, we demonstrate that the intracellular fibroblast growth factor homologous factors (FHF1-4) tune pathogenic I Na,L in an isoform-specific manner. This scheme suggests a complex orchestration of I Na,L in cardiomyocytes that may contribute to variable disease expressivity of NaV1.5 channelopathies. We further leverage these observations to engineer a peptide-inhibitor of I Na,L with a higher efficacy as compared to a well-established small-molecule inhibitor. Overall, these findings lend insights into molecular mechanisms underlying FHF regulation of I Na,L in pathophysiology and outline potential therapeutic avenues.
Collapse
Affiliation(s)
- Nourdine Chakouri
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Sharen Rivas
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Daniel Roybal
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Lin Yang
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Johanna Diaz
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Allen Hsu
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Ryan Mahling
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Bi-Xing Chen
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | | | - Deborah DiSilvestre
- Department Physiology, University of Maryland, Baltimore, MD, USA
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Dario Sirabella
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, NY, USA
| | - Barbara Corneo
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, NY, USA
| | - Gordon F. Tomaselli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ivy E. Dick
- Department Physiology, University of Maryland, Baltimore, MD, USA
| | - Steven O. Marx
- Department of Pharmacology, Columbia University, New York, NY, USA
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
15
|
Xiao Y, Theile JW, Zybura A, Pan Y, Lin Z, Cummins TR. A-type FHFs mediate resurgent currents through TTX-resistant voltage-gated sodium channels. eLife 2022; 11:77558. [PMID: 35441593 PMCID: PMC9071269 DOI: 10.7554/elife.77558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Resurgent currents (INaR) produced by voltage-gated sodium channels are required for many neurons to maintain high-frequency firing, and contribute to neuronal hyperexcitability and disease pathophysiology. Here we show, for the first time, that INaR can be reconstituted in a heterologous system by co-expression of sodium channel α-subunits and A-type fibroblast growth factor homologous factors (FHFs). Specifically, A-type FHFs induces INaR from Nav1.8, Nav1.9 tetrodotoxin-resistant neuronal channels and, to a lesser extent, neuronal Nav1.7 and cardiac Nav1.5 channels. Moreover, we identified the N-terminus of FHF as the critical molecule responsible for A-type FHFs-mediated INaR. Among the FHFs, FHF4A is the most important isoform for mediating Nav1.8 and Nav1.9 INaR. In nociceptive sensory neurons, FHF4A knockdown significantly reduces INaR amplitude and the percentage of neurons that generate INaR, substantially suppressing excitability. Thus, our work reveals a novel molecular mechanism underlying TTX-resistant INaR generation and provides important potential targets for pain treatment.
Collapse
Affiliation(s)
- Yucheng Xiao
- Biology Department, Indiana University - Purdue University Indianapolis, Indianapolis, United States
| | | | - Agnes Zybura
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, Indianapolis, United States
| | - Yanling Pan
- Biology Department, Indiana University - Purdue University Indianapolis, Indianapolis, United States
| | | | - Theodore R Cummins
- Biology Department, Indiana University - Purdue University Indianapolis, Indianapolis, United States
| |
Collapse
|
16
|
Narayanan DL, Majethia P, Shrikiran A, Siddiqui S, Dalal A, Shukla A. Further evidence of affected females with a heterozygous variant in FGF13 causing X-linked developmental and epileptic encephalopathy 90. Eur J Med Genet 2022; 65:104403. [PMID: 34871784 PMCID: PMC8918359 DOI: 10.1016/j.ejmg.2021.104403] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/18/2021] [Accepted: 11/28/2021] [Indexed: 01/03/2023]
Abstract
Developmental and epileptic encephalopathies (DEE) are a genetically heterogeneous group of disorders characterised by early onset epilepsy, epileptiform activity on electroencephalogram and associated developmental delay or neuroregression. With the advent of high throughput sequencing, novel gene-disease associations have been described for DEEs. Voltage activated sodium channels (Nav) regulate neuronal excitability. Fibroblast growth factor homologous factors (FHFs) are proteins, which bind to the C terminal cytoplasmic tails of alpha subunits of Nav channels and influence their function and surface expression. Gain of function hemizygous or heterozygous variants in FGF13 (also known as FHF2) were recently identified as the cause for X-linked developmental and epileptic encephalopathy 90 (DEE90; MIM# 301058) in seven individuals from five families, which included one female. We report an additional female, providing further evidence for a novel de novo heterozygous missense variant in FGF13, NM_004114.5: c.14T > G p.(Ile5Ser) causing X-linked DEE90. In addition, we review the genotype and phenotype of affected individuals with DEE90.
Collapse
Affiliation(s)
- Dhanya Lakshmi Narayanan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Purvi Majethia
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Aroor Shrikiran
- Department of Pediatrics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shahyan Siddiqui
- Department of Neuroimaging and Interventional Radiology, STAR Institute of Neurosciences, STAR Hospitals, Hyderabad, India
| | - Ashwin Dalal
- Diagnostics Division, Center for DNA Fingerprinting and Diagnostics, Hyderabad, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
17
|
Gao J, Xu G, Xu P. Whole-genome resequencing of three Coilia nasus population reveals genetic variations in genes related to immune, vision, migration, and osmoregulation. BMC Genomics 2021; 22:878. [PMID: 34872488 PMCID: PMC8647404 DOI: 10.1186/s12864-021-08182-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Background Coilia nasus is an important anadromous fish, widely distributed in China, Japan, and Korea. Based on morphological and ecological researches of C. nasus, two ecotypes were identified. One is the anadromous population (AP). The sexually mature fish run thousands of kilometers from marine to river for spawning. Another one is the resident population which cannot migrate. Based on their different habitats, they were classified into landlocked population (LP) and sea population (SP) which were resident in the freshwater lake and marine during the entire lifetime, respectively. However, they have never been systematically studied. Moreover, C. nasus is declining sharply due to overfishing and pollution recently. Therefore, further understandings of C. nasus populations are needed for germplasm protection. Results Whole-genome resequencing of AP, LP, and SP were performed to enrich the understanding of different populations of C. nasus. At the genome level, 3,176,204, 3,307,069, and 3,207,906 single nucleotide polymorphisms (SNPs) and 1,892,068, 2,002,912, and 1,922,168 insertion/deletion polymorphisms (InDels) were generated in AP, LP, and SP, respectively. Selective sweeping analysis showed that 1022 genes were selected in AP vs LP; 983 genes were selected in LP vs SP; 116 genes were selected in AP vs SP. Among them, selected genes related to immune, vision, migration, and osmoregulation were identified. Furthermore, their expression profiles were detected by quantitative real-time PCR. Expression levels of selected genes related to immune, and vision in LP were significantly lower than AP and SP. Selected genes related to migration in AP were expressed significantly more highly than LP. Expression levels of selected genes related to osmoregulation were also detected. The expression of NKAα and NKCC1 in LP were significantly lower than SP, while expression of NCC, SLC4A4, NHE3, and V-ATPase in LP was significantly higher than SP. Conclusions Combined to life history of C. nasus populations, our results revealed that the molecular mechanisms of their differences of immune, vision, migration, and osmoregulation. Our findings will provide a further understanding of different populations of C. nasus and will be beneficial for wild C. nasus protection. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08182-0.
Collapse
Affiliation(s)
- Jun Gao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, Jiangsu, China
| | - Gangchun Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, Jiangsu, China. .,Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, Jiangsu, China.
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, Jiangsu, China. .,Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, Jiangsu, China.
| |
Collapse
|
18
|
Li S, Chen LN, Zhu HJ, Feng X, Xie FY, Luo SM, Ou XH, Ma JY. Single-cell RNA sequencing analysis of mouse follicular somatic cells†. Biol Reprod 2021; 105:1234-1245. [PMID: 34467391 DOI: 10.1093/biolre/ioab163] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/29/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
Within the development of ovarian follicle, in addition to cell proliferation and differentiation, sophisticated cell-cell cross talks are established among follicular somatic cells such as granulosa cells (GCs) and theca cells. To systematically reveal the cell differentiation and signal transductions in follicular somatic cells, we collected the mouse follicular somatic cells from secondary to ovulatory stage, and analyzed the single cell transcriptomes. Having data filtered and screened, we found 6883 high variable genes in 4888 single cells. Then follicular somatic cells were clustered into 26 cell clusters, including 18 GC clusters, 4 theca endocrine cell (TEC) clusters, and 4 other somatic cell clusters, which include immune cells and Acta2 positive theca externa cells. From our data, we found there was metabolic reprogramming happened during GC differentiation. We also found both Cyp19a1 and Cyp11a1 could be expressed in TECs. We analyzed the expression patterns of genes associated with cell-cell interactions such as steroid hormone receptor genes, insulin signaling genes, and cytokine/transformation growth factor beta associated genes in all cell clusters. Lastly, we clustered the highly variable genes into 300 gene clusters, which could be used to search new genes involved in follicle development. These transcriptomes of follicular somatic cells provide us potential clues to reveal how mammals regulating follicle development and could help us find targets to improve oocyte quality for women with low fertility.
Collapse
Affiliation(s)
- Sen Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lei-Ning Chen
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hai-Jing Zhu
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,Teaching Center in Guangdong Second Provincial General Hospital, University of South China, Guangzhou, China
| | - Xie Feng
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Feng-Yun Xie
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shi-Ming Luo
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,Teaching Center in Guangdong Second Provincial General Hospital, University of South China, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jun-Yu Ma
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
19
|
Dvorak NM, Tapia CM, Baumgartner TJ, Singh J, Laezza F, Singh AK. Pharmacological Inhibition of Wee1 Kinase Selectively Modulates the Voltage-Gated Na + Channel 1.2 Macromolecular Complex. Cells 2021; 10:3103. [PMID: 34831326 PMCID: PMC8619224 DOI: 10.3390/cells10113103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Voltage-gated Na+ (Nav) channels are a primary molecular determinant of the action potential (AP). Despite the canonical role of the pore-forming α subunit in conferring this function, protein-protein interactions (PPI) between the Nav channel α subunit and its auxiliary proteins are necessary to reconstitute the full physiological activity of the channel and to fine-tune neuronal excitability. In the brain, the Nav channel isoforms 1.2 (Nav1.2) and 1.6 (Nav1.6) are enriched, and their activities are differentially regulated by the Nav channel auxiliary protein fibroblast growth factor 14 (FGF14). Despite the known regulation of neuronal Nav channel activity by FGF14, less is known about cellular signaling molecules that might modulate these regulatory effects of FGF14. To that end, and building upon our previous investigations suggesting that neuronal Nav channel activity is regulated by a kinase network involving GSK3, AKT, and Wee1, we interrogate in our current investigation how pharmacological inhibition of Wee1 kinase, a serine/threonine and tyrosine kinase that is a crucial component of the G2-M cell cycle checkpoint, affects the Nav1.2 and Nav1.6 channel macromolecular complexes. Our results show that the highly selective inhibitor of Wee1 kinase, called Wee1 inhibitor II, modulates FGF14:Nav1.2 complex assembly, but does not significantly affect FGF14:Nav1.6 complex assembly. These results are functionally recapitulated, as Wee1 inhibitor II entirely alters FGF14-mediated regulation of the Nav1.2 channel, but displays no effects on the Nav1.6 channel. At the molecular level, these effects of Wee1 inhibitor II on FGF14:Nav1.2 complex assembly and FGF14-mediated regulation of Nav1.2-mediated Na+ currents are shown to be dependent upon the presence of Y158 of FGF14, a residue known to be a prominent site for phosphorylation-mediated regulation of the protein. Overall, our data suggest that pharmacological inhibition of Wee1 confers selective modulatory effects on Nav1.2 channel activity, which has important implications for unraveling cellular signaling pathways that fine-tune neuronal excitability.
Collapse
Affiliation(s)
| | | | | | | | | | - Aditya K. Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 75901, USA; (N.M.D.); (C.M.T.); (T.J.B.); (J.S.); (F.L.)
| |
Collapse
|
20
|
Pan X, Zhao J, Zhou Z, Chen J, Yang Z, Wu Y, Bai M, Jiao Y, Yang Y, Hu X, Cheng T, Lu Q, Wang B, Li CL, Lu YJ, Diao L, Zhong YQ, Pan J, Zhu J, Xiao HS, Qiu ZL, Li J, Wang Z, Hui J, Bao L, Zhang X. 5'-UTR SNP of FGF13 causes translational defect and intellectual disability. eLife 2021; 10:63021. [PMID: 34184986 PMCID: PMC8241442 DOI: 10.7554/elife.63021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
The congenital intellectual disability (ID)-causing gene mutations remain largely unclear, although many genetic variations might relate to ID. We screened gene mutations in Chinese Han children suffering from severe ID and found a single-nucleotide polymorphism (SNP) in the 5′-untranslated region (5′-UTR) of fibroblast growth factor 13 (FGF13) mRNA (NM_001139500.1:c.-32c>G) shared by three male children. In both HEK293 cells and patient-derived induced pluripotent stem cells, this SNP reduced the translation of FGF13, which stabilizes microtubules in developing neurons. Mice carrying the homologous point mutation in 5′-UTR of Fgf13 showed delayed neuronal migration during cortical development, and weakened learning and memory. Furthermore, this SNP reduced the interaction between FGF13 5′-UTR and polypyrimidine-tract-binding protein 2 (PTBP2), which was required for FGF13 translation in cortical neurons. Thus, this 5′-UTR SNP of FGF13 interferes with the translational process of FGF13 and causes deficits in brain development and cognitive functions.
Collapse
Affiliation(s)
- Xingyu Pan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Jingrong Zhao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiying Zhou
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Jijun Chen
- Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Zhenxing Yang
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Yuxuan Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Meizhu Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yang Jiao
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yun Yang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xuye Hu
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Tianling Cheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianyun Lu
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bin Wang
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Chang-Lin Li
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Ying-Jin Lu
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Lei Diao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yan-Qing Zhong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Pan
- Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Jianmin Zhu
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Hua-Sheng Xiao
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Zi-Long Qiu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Zefeng Wang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Jingyi Hui
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Lan Bao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Xu Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
21
|
Zybura A, Hudmon A, Cummins TR. Distinctive Properties and Powerful Neuromodulation of Na v1.6 Sodium Channels Regulates Neuronal Excitability. Cells 2021; 10:1595. [PMID: 34202119 PMCID: PMC8307729 DOI: 10.3390/cells10071595] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) are critical determinants of cellular excitability. These ion channels exist as large heteromultimeric structures and their activity is tightly controlled. In neurons, the isoform Nav1.6 is highly enriched at the axon initial segment and nodes, making it critical for the initiation and propagation of neuronal impulses. Changes in Nav1.6 expression and function profoundly impact the input-output properties of neurons in normal and pathological conditions. While mutations in Nav1.6 may cause channel dysfunction, aberrant changes may also be the result of complex modes of regulation, including various protein-protein interactions and post-translational modifications, which can alter membrane excitability and neuronal firing properties. Despite decades of research, the complexities of Nav1.6 modulation in health and disease are still being determined. While some modulatory mechanisms have similar effects on other Nav isoforms, others are isoform-specific. Additionally, considerable progress has been made toward understanding how individual protein interactions and/or modifications affect Nav1.6 function. However, there is still more to be learned about how these different modes of modulation interact. Here, we examine the role of Nav1.6 in neuronal function and provide a thorough review of this channel's complex regulatory mechanisms and how they may contribute to neuromodulation.
Collapse
Affiliation(s)
- Agnes Zybura
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Andy Hudmon
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA;
| | - Theodore R. Cummins
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
22
|
Dvorak NM, Wadsworth PA, Wang P, Zhou J, Laezza F. Development of Allosteric Modulators of Voltage-Gated Na + Channels: A Novel Approach for an Old Target. Curr Top Med Chem 2021; 21:841-848. [PMID: 34036922 DOI: 10.2174/1568026621666210525105359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022]
Abstract
Given their primacy in governing the action potential (AP) of excitable cells, voltage-gated Na+ (Nav) channels are important pharmacological targets of therapeutics for a diverse array of clinical indications. Despite historically being a traditional drug target, therapeutics targeting Nav channels lack isoform selectivity, giving rise to off-target side effects. To develop isoform-selective modulators of Nav channels with improved target-specificity, the identification and pharmacological targeting of allosteric sites that display structural divergence among Nav channel isoforms represents an attractive approach. Despite the high homology among Nav channel α subunit isoforms (Nav1.1-Nav1.9), there is considerable amino acid sequence divergence among their constituent C-terminal domains (CTD), which enables structurally and functionally specific protein: protein interactions (PPI) with auxiliary proteins. Although pharmacological targeting of such PPI interfaces between the CTDs of Nav channels and auxiliary proteins represents an innovate approach for developing isoform-selective modulators of Nav channels, appreciable modulation of PPIs using small molecules has conventionally been difficult to achieve. After briefly discussing the challenges of modulating PPIs using small molecules, this current frontier review that follows subsequently expounds on approaches for circumventing such difficulties in the context of developing small molecule modulators of PPIs between transmembrane ion channels and their auxiliary proteins. In addition to broadly discussing such approaches, the implementation of such approaches is specifically discussed in the context of developing small molecule modulators between the CTD of Nav channels and auxiliary proteins. Developing allosteric modulators of ion channels by targeting their PPI interfaces with auxiliary proteins represents an innovative and promising strategy in ion channel drug discovery that could expand the "druggable genome" and usher in first-in-class PPI-targeting therapeutics for a multitude of channelopathies.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Paul A Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| |
Collapse
|
23
|
Yang QQ, Zhai YQ, Wang HF, Cai YC, Ma XY, Yin YQ, Li YD, Zhou GM, Zhang X, Hu G, Zhou JW. Nuclear isoform of FGF13 regulates post-natal neurogenesis in the hippocampus through an epigenomic mechanism. Cell Rep 2021; 35:109127. [PMID: 34010636 DOI: 10.1016/j.celrep.2021.109127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 02/13/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022] Open
Abstract
The hippocampus is one of two niches in the mammalian brain with persistent neurogenesis into adulthood. The neurogenic capacity of hippocampal neural stem cells (NSCs) declines with age, but the molecular mechanisms of this process remain unknown. In this study, we find that fibroblast growth factor 13 (FGF13) is essential for the post-natal neurogenesis in mouse hippocampus, and FGF13 deficiency impairs learning and memory. In particular, we find that FGF13A, the nuclear isoform of FGF13, is involved in the maintenance of NSCs and the suppression of neuronal differentiation during post-natal hippocampal development. Furthermore, we find that FGF13A interacts with ARID1B, a unit of Brahma-associated factor chromatin remodeling complex, and suppresses the expression of neuron differentiation-associated genes through chromatin modification. Our results suggest that FGF13A is an important regulator for maintaining the self-renewal and neurogenic capacity of NSCs in post-natal hippocampus, revealing an epigenomic regulatory function of FGFs in neurogenesis.
Collapse
Affiliation(s)
- Qiao-Qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Ying-Qi Zhai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hai-Fang Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Chen Cai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin-Yue Ma
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Yan-Qing Yin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan-Dong Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guo-Min Zhou
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Jia-Wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China; Co-innovation Center of Neuroregeneration, School of Medicine, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
24
|
Martinez-Espinosa PL, Neely A, Ding J, Lingle CJ. Fast inactivation of Nav current in rat adrenal chromaffin cells involves two independent inactivation pathways. J Gen Physiol 2021; 153:211834. [PMID: 33647101 PMCID: PMC7927663 DOI: 10.1085/jgp.202012784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Voltage-dependent sodium (Nav) current in adrenal chromaffin cells (CCs) is rapidly inactivating and tetrodotoxin (TTX)–sensitive. The fractional availability of CC Nav current has been implicated in regulation of action potential (AP) frequency and the occurrence of slow-wave burst firing. Here, through recordings of Nav current in rat CCs, primarily in adrenal medullary slices, we describe unique inactivation properties of CC Nav inactivation that help define AP firing rates in CCs. The key feature of CC Nav current is that recovery from inactivation, even following brief (5 ms) inactivation steps, exhibits two exponential components of similar amplitude. Various paired pulse protocols show that entry into the fast and slower recovery processes result from largely independent competing inactivation pathways, each of which occurs with similar onset times at depolarizing potentials. Over voltages from −120 to −80 mV, faster recovery varies from ∼3 to 30 ms, while slower recovery varies from ∼50 to 400 ms. With strong depolarization (above −10 mV), the relative entry into slow or fast recovery pathways is similar and independent of voltage. Trains of short depolarizations favor recovery from fast recovery pathways and result in cumulative increases in the slow recovery fraction. Dual-pathway fast inactivation, by promoting use-dependent accumulation in slow recovery pathways, dynamically regulates Nav availability. Consistent with this finding, repetitive AP clamp waveforms at 1–10 Hz frequencies reduce Nav availability 80–90%, depending on holding potential. These results indicate that there are two distinct pathways of fast inactivation, one leading to conventional fast recovery and the other to slower recovery, which together are well-suited to mediate use-dependent changes in Nav availability.
Collapse
Affiliation(s)
| | - Alan Neely
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Jiuping Ding
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
25
|
Fry AE, Marra C, Derrick AV, Pickrell WO, Higgins AT, Te Water Naude J, McClatchey MA, Davies SJ, Metcalfe KA, Tan HJ, Mohanraj R, Avula S, Williams D, Brady LI, Mesterman R, Tarnopolsky MA, Zhang Y, Yang Y, Wang X, Rees MI, Goldfarb M, Chung SK. Missense variants in the N-terminal domain of the A isoform of FHF2/FGF13 cause an X-linked developmental and epileptic encephalopathy. Am J Hum Genet 2021; 108:176-185. [PMID: 33245860 PMCID: PMC7820623 DOI: 10.1016/j.ajhg.2020.10.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/30/2020] [Indexed: 01/22/2023] Open
Abstract
Fibroblast growth factor homologous factors (FHFs) are intracellular proteins which regulate voltage-gated sodium (Nav) channels in the brain and other tissues. FHF dysfunction has been linked to neurological disorders including epilepsy. Here, we describe two sibling pairs and three unrelated males who presented in infancy with intractable focal seizures and severe developmental delay. Whole-exome sequencing identified hemi- and heterozygous variants in the N-terminal domain of the A isoform of FHF2 (FHF2A). The X-linked FHF2 gene (also known as FGF13) has alternative first exons which produce multiple protein isoforms that differ in their N-terminal sequence. The variants were located at highly conserved residues in the FHF2A inactivation particle that competes with the intrinsic fast inactivation mechanism of Nav channels. Functional characterization of mutant FHF2A co-expressed with wild-type Nav1.6 (SCN8A) revealed that mutant FHF2A proteins lost the ability to induce rapid-onset, long-term blockade of the channel while retaining pro-excitatory properties. These gain-of-function effects are likely to increase neuronal excitability consistent with the epileptic potential of FHF2 variants. Our findings demonstrate that FHF2 variants are a cause of infantile-onset developmental and epileptic encephalopathy and underline the critical role of the FHF2A isoform in regulating Nav channel function.
Collapse
Affiliation(s)
- Andrew E Fry
- Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK; Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Christopher Marra
- Department of Biological Sciences, Hunter College of City University, 695 Park Avenue, New York, NY 10065, USA; Program in Biology, Graduate Center of City University, 365 Fifth Avenue, New York, NY 10016, USA
| | - Anna V Derrick
- Neurology and Molecular Neuroscience Research, Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - William O Pickrell
- Neurology and Molecular Neuroscience Research, Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK; Neurology department, Morriston Hospital, Swansea Bay University Hospital Health Board, Swansea SA6 6NL, UK
| | - Adam T Higgins
- Neurology and Molecular Neuroscience Research, Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Johann Te Water Naude
- Paediatric Neurology, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Martin A McClatchey
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Sally J Davies
- Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Kay A Metcalfe
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust and Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
| | - Hui Jeen Tan
- Department of Paediatric Neurology, Royal Manchester Children's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Rajiv Mohanraj
- Department of Neurology, Salford Royal Hospital NHS Foundation Trust, Stott Lane, Salford M6 8HD, UK
| | - Shivaram Avula
- Department of Radiology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool L12 2AP, UK
| | - Denise Williams
- West Midlands Regional Genetics Service, Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - Lauren I Brady
- Department of Paediatrics, McMaster University, 1200 Main St. W., Hamilton, ON L8N 3Z5, Canada
| | - Ronit Mesterman
- Department of Paediatrics, McMaster University, 1200 Main St. W., Hamilton, ON L8N 3Z5, Canada
| | - Mark A Tarnopolsky
- Department of Paediatrics, McMaster University, 1200 Main St. W., Hamilton, ON L8N 3Z5, Canada
| | - Yuehua Zhang
- Department of Pediatrics, Peking University First Hospital, Xicheng District, Beijing 100034, China
| | - Ying Yang
- Department of Pediatrics, Peking University First Hospital, Xicheng District, Beijing 100034, China
| | | | - Mark I Rees
- Neurology and Molecular Neuroscience Research, Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK; Faculty of Medicine and Health, Camperdown, University of Sydney, NSW 2006, Australia
| | - Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, 695 Park Avenue, New York, NY 10065, USA; Program in Biology, Graduate Center of City University, 365 Fifth Avenue, New York, NY 10016, USA
| | - Seo-Kyung Chung
- Neurology and Molecular Neuroscience Research, Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK; Kids Neuroscience Centre, Kids Research, Children Hospital at Westmead, Sydney, NSW 2145, Australia; Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, NSW 2050, Australia
| |
Collapse
|
26
|
Wang P, Wadsworth PA, Dvorak NM, Singh AK, Chen H, Liu Z, Zhou R, Holthauzen LMF, Zhou J, Laezza F. Design, Synthesis, and Pharmacological Evaluation of Analogues Derived from the PLEV Tetrapeptide as Protein-Protein Interaction Modulators of Voltage-Gated Sodium Channel 1.6. J Med Chem 2020; 63:11522-11547. [PMID: 33054193 DOI: 10.1021/acs.jmedchem.0c00531] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The voltage-gated Na+ (Nav) channel is the molecular determinant of excitability. Disruption of protein-protein interactions (PPIs) between Nav1.6 and fibroblast growth factor 14 (FGF14) leads to impaired excitability of neurons in clinically relevant brain areas associated with channelopathies. Here, we designed, synthesized, and pharmacologically characterized new peptidomimetics based on a PLEV tetrapeptide scaffold derived from the FGF14:Nav1.6 PPI interface. Addition of an N-terminal 1-adamantanecarbonyl pharmacophore significantly improved peptidomimetic inhibitory potency. Surface plasmon resonance studies revealed that while this moiety was sufficient to confer binding to FGF14, altering the C-terminal moiety from methoxy (21a) to π bond-containing (23a and 23b) or cycloalkane substituents (23e) abrogated the binding to Nav1.6. Whole-cell patch-clamp electrophysiology subsequently revealed that 21a had functionally relevant interactions with both the C-terminal tail of Nav1.6 and FGF14. Collectively, these findings support that 21a (PW0564) may serve as a promising lead to develop target-selective neurotherapeutics by modulating protein-channel interactions.
Collapse
|
27
|
Menezes LFS, Sabiá Júnior EF, Tibery DV, Carneiro LDA, Schwartz EF. Epilepsy-Related Voltage-Gated Sodium Channelopathies: A Review. Front Pharmacol 2020; 11:1276. [PMID: 33013363 PMCID: PMC7461817 DOI: 10.3389/fphar.2020.01276] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
Epilepsy is a disease characterized by abnormal brain activity and a predisposition to generate epileptic seizures, leading to neurobiological, cognitive, psychological, social, and economic impacts for the patient. There are several known causes for epilepsy; one of them is the malfunction of ion channels, resulting from mutations. Voltage-gated sodium channels (NaV) play an essential role in the generation and propagation of action potential, and malfunction caused by mutations can induce irregular neuronal activity. That said, several genetic variations in NaV channels have been described and associated with epilepsy. These mutations can affect channel kinetics, modifying channel activation, inactivation, recovery from inactivation, and/or the current window. Among the NaV subtypes related to epilepsy, NaV1.1 is doubtless the most relevant, with more than 1500 mutations described. Truncation and missense mutations are the most observed alterations. In addition, several studies have already related mutated NaV channels with the electrophysiological functioning of the channel, aiming to correlate with the epilepsy phenotype. The present review provides an overview of studies on epilepsy-associated mutated human NaV1.1, NaV1.2, NaV1.3, NaV1.6, and NaV1.7.
Collapse
Affiliation(s)
- Luis Felipe Santos Menezes
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Elias Ferreira Sabiá Júnior
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Lilian Dos Anjos Carneiro
- Faculdade de Medicina, Centro Universitário Euro Americano, Brasília, Brazil.,Faculdade de Medicina, Centro Universitário do Planalto Central, Brasília, Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
28
|
Singh AK, Wadsworth PA, Tapia CM, Aceto G, Ali SR, Chen H, D'Ascenzo M, Zhou J, Laezza F. Mapping of the FGF14:Nav1.6 complex interface reveals FLPK as a functionally active peptide modulating excitability. Physiol Rep 2020; 8:e14505. [PMID: 32671946 PMCID: PMC7363588 DOI: 10.14814/phy2.14505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
The voltage-gated sodium (Nav) channel complex is comprised of pore-forming α subunits (Nav1.1-1.9) and accessory regulatory proteins such as the intracellular fibroblast growth factor 14 (FGF14). The cytosolic Nav1.6 C-terminal tail binds directly to FGF14 and this interaction modifies Nav1.6-mediated currents with effects on intrinsic excitability in the brain. Previous studies have identified the FGF14V160 residue within the FGF14 core domain as a hotspot for the FGF14:Nav1.6 complex formation. Here, we used three short amino acid peptides around FGF14V160 to probe for the FGF14 interaction with the Nav1.6 C-terminal tail and to evaluate the activity of the peptide on Nav1.6-mediated currents. In silico docking predicts FLPK to bind to FGF14V160 with the expectation of interfering with the FGF14:Nav1.6 complex formation, a phenotype that was confirmed by the split-luciferase assay (LCA) and surface plasmon resonance (SPR), respectively. Whole-cell patch-clamp electrophysiology studies demonstrate that FLPK is able to prevent previously reported FGF14-dependent phenotypes of Nav1.6 currents, but that its activity requires the FGF14 N-terminal tail, a domain that has been shown to contribute to Nav1.6 inactivation independently from the FGF14 core domain. In medium spiny neurons in the nucleus accumbens, where both FGF14 and Nav1.6 are abundantly expressed, FLPK significantly increased firing frequency by a mechanism consistent with the ability of the tetrapeptide to interfere with Nav1.6 inactivation and potentiate persistent Na+ currents. Taken together, these results indicate that FLPK might serve as a probe for characterizing molecular determinants of neuronal excitability and a peptide scaffold to develop allosteric modulators of Nav channels.
Collapse
Affiliation(s)
- Aditya K. Singh
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Paul A. Wadsworth
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- M.D.‐Ph.D. Combined Degree ProgramUniversità Cattolica del Sacro CuoreRomeItaly
- Biochemistry and Molecular Biology Graduate ProgramUniversità Cattolica del Sacro CuoreRomeItaly
| | - Cynthia M. Tapia
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- NIEHS Environmental Toxicology Training ProgramUniversità Cattolica del Sacro CuoreRomeItaly
| | - Giuseppe Aceto
- Institute of Human PhysiologyUniversità Cattolica del Sacro CuoreRomeItaly
- Department of NeuroscienceUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario A. GemelliIRCCSRomeItaly
| | - Syed R. Ali
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Haiying Chen
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Marcello D'Ascenzo
- Institute of Human PhysiologyUniversità Cattolica del Sacro CuoreRomeItaly
- Department of NeuroscienceUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario A. GemelliIRCCSRomeItaly
| | - Jia Zhou
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
| | - Fernanda Laezza
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
- Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
29
|
Navarro MA, Salari A, Lin JL, Cowan LM, Penington NJ, Milescu M, Milescu LS. Sodium channels implement a molecular leaky integrator that detects action potentials and regulates neuronal firing. eLife 2020; 9:54940. [PMID: 32101161 PMCID: PMC7043890 DOI: 10.7554/elife.54940] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/13/2020] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated sodium channels play a critical role in cellular excitability, amplifying small membrane depolarizations into action potentials. Interactions with auxiliary subunits and other factors modify the intrinsic kinetic mechanism to result in new molecular and cellular functionality. We show here that sodium channels can implement a molecular leaky integrator, where the input signal is the membrane potential and the output is the occupancy of a long-term inactivated state. Through this mechanism, sodium channels effectively measure the frequency of action potentials and convert it into Na+ current availability. In turn, the Na+ current can control neuronal firing frequency in a negative feedback loop. Consequently, neurons become less sensitive to changes in excitatory input and maintain a lower firing rate. We present these ideas in the context of rat serotonergic raphe neurons, which fire spontaneously at low frequency and provide critical neuromodulation to many autonomous and cognitive brain functions.
Collapse
Affiliation(s)
- Marco A Navarro
- Division of Biological Sciences, University of Missouri, Columbia, United States
| | - Autoosa Salari
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Jenna L Lin
- Division of Biological Sciences, University of Missouri, Columbia, United States
| | - Luke M Cowan
- Division of Biological Sciences, University of Missouri, Columbia, United States
| | - Nicholas J Penington
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, United States
| | - Mirela Milescu
- Division of Biological Sciences, University of Missouri, Columbia, United States
| | - Lorin S Milescu
- Division of Biological Sciences, University of Missouri, Columbia, United States.,Department of Biology, University of Maryland, College Park, United States
| |
Collapse
|
30
|
Li Q, Zhai Z, Li J. Fibroblast growth factor homologous factors are potential ion channel modifiers associated with cardiac arrhythmias. Eur J Pharmacol 2020; 871:172920. [PMID: 31935396 DOI: 10.1016/j.ejphar.2020.172920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/10/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022]
Abstract
Stable electrical activity in cardiac myocytes is the basis of maintaining normal myocardial systolic and diastolic function. Cardiac ionic currents and their associated regulatory proteins are crucial to myocyte excitability and heart function. Fibroblast growth factor homologous factors (FHFs) are intracellular noncanonical fibroblast growth factors (FGFs) that are incapable of activating FGF receptors. The main functions of FHFs are to regulate ion channels and influence excitability, which are processes involved in sustaining normal cardiac function. In addition to their regulatory effect on ion channels, FHFs can be regulators of cardiac hypertrophic signaling and alter signaling pathways, including the protein kinase, NF<kappa>B, and p53 pathways, which are related to the pathological processes of heart diseases. This review emphasizes FHF-mediated regulation of cardiac excitability and the association of FHFs with cardiac arrhythmias and explores the idea that abnormal FHFs may be an unrecognized cause of cardiac disorders.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhenyu Zhai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
31
|
Sinden DS, Holman CD, Bare CJ, Sun X, Gade AR, Cohen DE, Pitt GS. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity. FASEB J 2019; 33:11579-11594. [PMID: 31339804 PMCID: PMC6994920 DOI: 10.1096/fj.201901178r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor (FGF)13, a nonsecreted, X-linked, FGF homologous factor, is differentially expressed in adipocytes in response to diet, yet Fgf13's role in metabolism has not been explored. Heterozygous Fgf13 knockouts fed normal chow and housed at 22°C showed hyperactivity accompanying reduced core temperature and obesity when housed at 30°C. Those heterozygous knockouts showed defects in thermogenesis even at 30°C and an inability to protect core temperature. Surprisingly, we detected trivial FGF13 in adipose of wild-type mice fed normal chow and no obesity in adipose-specific heterozygous knockouts housed at 30°C, and we detected an intact brown fat response through exogenous β3 agonist stimulation, suggesting a defect in sympathetic drive to brown adipose tissue. In contrast, hypothalamic-specific ablation of Fgf13 recapitulated weight gain at 30°C. Norepinephrine turnover in brown fat was reduced at both housing temperatures. Thus, our data suggest that impaired CNS regulation of sympathetic activation of brown fat underlies obesity and thermogenesis in Fgf13 heterozygous knockouts fed normal chow.-Sinden, D. S., Holman, C. D., Bare, C. J., Sun, X., Gade, A. R., Cohen, D. E., Pitt, G. S. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity.
Collapse
Affiliation(s)
- Daniel S. Sinden
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Corey D. Holman
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Curtis J. Bare
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Xiaolu Sun
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Aravind R. Gade
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
32
|
Effraim PR, Huang J, Lampert A, Stamboulian S, Zhao P, Black JA, Dib-Hajj SD, Waxman SG. Fibroblast growth factor homologous factor 2 (FGF-13) associates with Nav1.7 in DRG neurons and alters its current properties in an isoform-dependent manner. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100029. [PMID: 31223136 PMCID: PMC6565799 DOI: 10.1016/j.ynpai.2019.100029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022]
Abstract
Fibroblast Growth Factor Homologous Factors (FHF) constitute a subfamily of FGF proteins with four prototypes (FHF1-4; also known as FGF11-14). FHF proteins have been shown to bind directly to the membrane-proximal segment of the C-terminus in voltage-gated sodium channels (Nav), and regulate current density, availability, and frequency-dependent inhibition of sodium currents. Members of the FHF2 subfamily, FHF2A and FHF2B, differ in the length and sequence of their N-termini, and, importantly, differentially regulate Nav1.6 gating properties. Using immunohistochemistry, we show that FHF2 isoforms are expressed in adult dorsal root ganglion (DRG) neurons where they co-localize with Nav1.6 and Nav1.7. FHF2A and FHF2B show differential localization in neuronal compartments in DRG neurons, and levels of expression of FHF2 factors are down-regulated following sciatic nerve axotomy. Because Nav1.7 in nociceptors plays a critical role in pain, we reasoned that its interaction with FHF2 isoforms might regulate its current properties. Using whole-cell patch clamp in heterologous expression systems, we show that the expression of FHF2A in HEK293 cell line stably expressing Nav1.7 channels causes no change in activation, whereas FHF2B depolarizes activation. Both FHF2 isoforms depolarize fast-inactivation. Additionally, FHF2A causes an accumulation of inactivated channels at all frequencies tested due to a slowing of recovery from inactivation, whereas FHF2B has little effect on these properties of Nav1.7. Measurements of the Nav1.7 current in DRG neurons in which FHF2 levels are knocked down confirmed the effects of FHF2A on repriming, and FHF2B on activation, however FHF2A and B did not have an effect on fast inactivation. Our data demonstrates that FHF2 does indeed regulate the current properties of Nav1.7 and does so in an isoform and cell-specific manner.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Severine Stamboulian
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Joel A. Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
33
|
Mo J, Chen X, Ni C, Wu K, Li X, Zhu Q, Ma L, Chen Y, Zhang S, Wang Y, Lian Q, Ge RS. Fibroblast growth factor homologous factor 1 stimulates Leydig cell regeneration from stem cells in male rats. J Cell Mol Med 2019; 23:5618-5631. [PMID: 31222931 PMCID: PMC6653537 DOI: 10.1111/jcmm.14461] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 01/05/2023] Open
Abstract
Fibroblast growth factor homologous factor 1 (FHF1) is an intracellular protein that does not bind to cell surface fibroblast growth factor receptor. Here, we report that FHF1 is abundantly present in Leydig cells with up‐regulation during its development. Adult male Sprague Dawley rats were intraperitoneally injected with 75 mg/kg ethane dimethane sulphonate (EDS) to ablate Leydig cells to initiate their regeneration. Then, rats daily received intratesticular injection of FHF1 (0, 10 and 100 ng/testis) from post‐EDS day 14 for 14 days. FHF1 increased serum testosterone levels without affecting the levels of luteinizing hormone and follicle‐stimulating hormone. FHF1 increased the cell number staining with HSD11B1, a biomarker for Leydig cells at the advanced stage, without affecting the cell number staining with CYP11A1, a biomarker for all Leydig cells. FHF1 did not affect PCNA‐labelling index in Leydig cells. FHF1 increased Leydig cell mRNA (Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Insl3, Nr5a1 and Hsd11b1) and their protein levels in vivo. FHF1 increased preadipocyte biomarker Dlk1 mRNA level and decreased fully differentiated adipocyte biomarker (Fabp4 and Lpl) mRNA and their protein levels. In conclusion, FHF1 promotes Leydig cell regeneration from stem cells while inhibiting the differentiation of preadipocyte/stem cells into adipocytes in EDS‐treated testis.
Collapse
Affiliation(s)
- Jiaying Mo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuxiu Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaobo Ni
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Keyang Wu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoheng Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiqi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Leika Ma
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yong Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Song Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyan Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingquan Lian
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Israel MR, Tanaka BS, Castro J, Thongyoo P, Robinson SD, Zhao P, Deuis JR, Craik DJ, Durek T, Brierley SM, Waxman SG, Dib-Hajj SD, Vetter I. Na V 1.6 regulates excitability of mechanosensitive sensory neurons. J Physiol 2019; 597:3751-3768. [PMID: 31087362 DOI: 10.1113/jp278148] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The β-scorpion toxin, Cn2, is selective for NaV 1.6 in dorsal root ganglion neurons. NaV 1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. NaV 1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted NaV isoforms in pain generation. ABSTRACT Peripheral sensory neurons express multiple voltage-gated sodium channels (NaV ) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): NaV 1.7, NaV 1.8 and NaV 1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests NaV 1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of NaV 1.6 channels in peripheral sensory neurons. The potent and NaV 1.6 selective β-scorpion toxin Cn2 was used to assess the effect of NaV 1.6 channel activation in the PNS. The multidisciplinary approach included Ca2+ imaging, whole-cell patch-clamp recordings, skin-nerve and gut-nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates NaV 1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, NaV 1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on NaV 1.6 channels in sensory neurons. Activation of NaV 1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing NaV 1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.
Collapse
Affiliation(s)
- Mathilde R Israel
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Brian S Tanaka
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Joel Castro
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Panumart Thongyoo
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Samuel D Robinson
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jennifer R Deuis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David J Craik
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Thomas Durek
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Irina Vetter
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
35
|
Ransdell JL, Nerbonne JM. Voltage-gated sodium currents in cerebellar Purkinje neurons: functional and molecular diversity. Cell Mol Life Sci 2018; 75:3495-3505. [PMID: 29982847 PMCID: PMC6123253 DOI: 10.1007/s00018-018-2868-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
Purkinje neurons, the sole output of the cerebellar cortex, deliver GABA-mediated inhibition to the deep cerebellar nuclei. To subserve this critical function, Purkinje neurons fire repetitively, and at high frequencies, features that have been linked to the unique properties of the voltage-gated sodium (Nav) channels expressed. In addition to the rapidly activating and inactivating, or transient, component of the Nav current (INaT) present in many types of central and peripheral neurons, Purkinje neurons, also expresses persistent (INaP) and resurgent (INaR) Nav currents. Considerable progress has been made in detailing the biophysical properties and identifying the molecular determinants of these discrete Nav current components, as well as defining their roles in the regulation of Purkinje neuron excitability. Here, we review this important work and highlight the remaining questions about the molecular mechanisms controlling the expression and the functioning of Nav currents in Purkinje neurons. We also discuss the impact of the dynamic regulation of Nav currents on the functioning of individual Purkinje neurons and cerebellar circuits.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Jeanne M Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
| |
Collapse
|
36
|
Hoxha E, Marcinnò A, Montarolo F, Masante L, Balbo I, Ravera F, Laezza F, Tempia F. Emerging roles of Fgf14 in behavioral control. Behav Brain Res 2018; 356:257-265. [PMID: 30189289 PMCID: PMC10082543 DOI: 10.1016/j.bbr.2018.08.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/03/2018] [Accepted: 08/31/2018] [Indexed: 01/19/2023]
Abstract
Sexual disturbances, and aggressivity are a major social problem. However, the molecular mechanisms involved in the control of these behaviors are largely unknown. FGF14, which is an intracellular protein controlling neuronal excitability and synaptic transmission, has been implied in neurologic and psychiatric disorders. Mice with Fgf14 deletion show blunted responses to drugs of abuse. By behavioral tests we show that male Fgf14 knockout mice have a marked reduction of several behaviors including aggressivity and sexual behavior. Other behaviors driven by spontaneous initiative like burying novel objects and spontaneous digging and climbing are also reduced in Fgf14 knockout mice. These deficits cannot be attributed to a generalized decrease of activity levels, because in the open field test Fgf14 knockout mice have the same spontaneous locomotion as wild types and increased rearing. Our results show that Fgf14 is important to preserve a set of behaviors and suggest that fine tuning of neuronal function by Fgf14 is an important mechanism of control for such behaviors.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy; Department of Neuroscience, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Andrea Marcinnò
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Linda Masante
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy; Department of Neuroscience, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Francesco Ravera
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy; Department of Neuroscience, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043 Orbassano, Italy; Department of Neuroscience, University of Torino, Corso Raffaello 30, 10125, Torino, Italy; National Neuroscience Institute (Italy), Corso Massimo D'Azeglio 52, 10126 Torino, Italy.
| |
Collapse
|
37
|
Fibroblast growth factor 12 is expressed in spiral and vestibular ganglia and necessary for auditory and equilibrium function. Sci Rep 2018; 8:11491. [PMID: 30065296 PMCID: PMC6068167 DOI: 10.1038/s41598-018-28618-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/26/2018] [Indexed: 11/13/2022] Open
Abstract
We investigated fibroblast growth factor 12 (FGF12) as a transcript enriched in the inner ear by searching published cDNA library databases. FGF12 is a fibroblast growth factor homologous factor, a subset of the FGF superfamily. To date, its localisation and function in the inner ear have not been determined. Here, we show that FGF12 mRNA is localised in spiral ganglion neurons (SGNs) and the vestibular ganglion. We also show that FGF12 protein is localised in SGNs, the vestibular ganglion, and nerve fibres extending beneath hair cells. Moreover, we investigated FGF12 function in auditory and vestibular systems using Fgf12-knockout (FGF12-KO) mice generated with CRISPR/Cas9 technology. Our results show that the inner ear morphology of FGF12-KO mice is not significantly different compared with wild-type mice. However, FGF12-KO mice exhibited an increased hearing threshold, as measured by the auditory brainstem response, as well as deficits in rotarod and balance beam performance tests. These results suggest that FGF12 is necessary for normal auditory and equilibrium function.
Collapse
|
38
|
Dash B, Han C, Waxman SG, Dib-Hajj SD. Nonmuscle myosin II isoforms interact with sodium channel alpha subunits. Mol Pain 2018; 14:1744806918788638. [PMID: 29956586 PMCID: PMC6052497 DOI: 10.1177/1744806918788638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Sodium channels play pivotal roles in health and diseases due to their ability to control cellular excitability. The pore-forming α-subunits (sodium channel alpha subunits) of the voltage-sensitive channels (i.e., Nav1.1–1.9) and the nonvoltage-dependent channel (i.e., Nax) share a common structural motif and selectivity for sodium ions. We hypothesized that the actin-based nonmuscle myosin II motor proteins, nonmuscle myosin heavy chain-IIA/myh9, and nonmuscle myosin heavy chain-IIB/myh10 might interact with sodium channel alpha subunits to play an important role in their transport, trafficking, and/or function. Immunochemical and electrophysiological assays were conducted using rodent nervous (brain and dorsal root ganglia) tissues and ND7/23 cells coexpressing Nav subunits and recombinant myosins. Immunoprecipitation of myh9 and myh10 from rodent brain tissues led to the coimmunoprecipitation of Nax, Nav1.2, and Nav1.3 subunits, but not Nav1.1 and Nav1.6 subunits, expressed there. Similarly, immunoprecipitation of myh9 and myh10 from rodent dorsal root ganglia tissues led to the coimmunoprecipitation of Nav1.7 and Nav1.8 subunits, but not Nav1.9 subunits, expressed there. The functional implication of one of these interactions was assessed by coexpressing myh10 along with Nav1.8 subunits in ND7/23 cells. Myh10 overexpression led to three-fold increase (P < 0.01) in the current density of Nav1.8 channels expressed in ND7/23 cells. Myh10 coexpression also hyperpolarized voltage-dependent activation and steady-state fast inactivation of Nav1.8 channels. In addition, coexpression of myh10 reduced (P < 0.01) the offset of fast inactivation and the amplitude of the ramp currents of Nav1.8 channels. These results indicate that nonmuscle myosin heavy chain-IIs interact with sodium channel alpha subunits subunits in an isoform-dependent manner and influence their functional properties.
Collapse
Affiliation(s)
- Bhagirathi Dash
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Chongyang Han
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Sulayman D Dib-Hajj
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
39
|
Heide R, Bostock H, Ventzel L, Grafe P, Bergmans J, Fuglsang-Frederiksen A, Finnerup NB, Tankisi H. Axonal excitability changes and acute symptoms of oxaliplatin treatment: In vivo evidence for slowed sodium channel inactivation. Clin Neurophysiol 2017; 129:694-706. [PMID: 29233604 DOI: 10.1016/j.clinph.2017.11.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 10/16/2017] [Accepted: 11/05/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Neurotoxicity is the most frequent dose-limiting side effect of the anti-cancer agent oxaliplatin, but the mechanisms are not well understood. This study used nerve excitability testing to investigate the pathophysiology of the acute neurotoxicity. METHODS Questionnaires, quantitative sensory tests, nerve conduction studies and nerve excitability testing were undertaken in 12 patients with high-risk colorectal cancer treated with adjuvant oxaliplatin and in 16 sex- and age-matched healthy controls. Examinations were performed twice for patients: once within 3 days after oxaliplatin treatment (post-infusion examination) and once shortly before the following treatment (recovery examination). RESULTS The most frequent post-infusion symptoms were tingling paresthesias and cold allodynia. The most prominent nerve excitability change was decreased superexcitability of motor axons which correlated with the average intensity of abnormal sensations (Spearman Rho = 0.80, p < .01). The motor nerve excitability changes were well modeled by a slowing of sodium channel inactivation, and were proportional to dose/m2 with a half-life of about 10d. CONCLUSIONS Oxaliplatin induces reversible slowing of sodium channel inactivation in motor axons, and these changes are closely related to the reversible cold allodynia. However, further studies are required due to small sample size in this study. SIGNIFICANCE Nerve excitability data provide an index of sodium channel dysfunction: an objective biomarker of acute oxaliplatin neurotoxicity.
Collapse
Affiliation(s)
- Rikke Heide
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark; Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hugh Bostock
- Institute of Neurology, Queen Square House, London, United Kingdom
| | - Lise Ventzel
- Department of Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Grafe
- Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joseph Bergmans
- Laboratory of Clinical Neurophysiology, Faculty of Medicine, University of Louvain, Brussels, Belgium
| | | | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
40
|
Otani Y, Ichikawa T, Kurozumi K, Inoue S, Ishida J, Oka T, Shimizu T, Tomita Y, Hattori Y, Uneda A, Matsumoto Y, Michiue H, Date I. Fibroblast growth factor 13 regulates glioma cell invasion and is important for bevacizumab-induced glioma invasion. Oncogene 2017; 37:777-786. [PMID: 29059154 DOI: 10.1038/onc.2017.373] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 12/16/2022]
Abstract
Glioblastoma has the poorest prognosis, and is characterized by excessive invasion and angiogenesis. To determine the invasive mechanisms, we previously used two glioma cell lines (J3T-1 and J3T-2) with different invasive phenotypes. The J3T-1 showed abundant angiogenesis and tumor cell invasion around neovasculature, while J3T-2 showed diffuse cell infiltration into surrounding healthy parenchyma. Microarray analyses were used to identify invasion-related genes in J3T-2 cells, and the expressed genes and their intracellular and intratumoral distribution patterns were evaluated in J3T-2 cell lines, human glioma cell lines, human glioblastoma stem cells and human glioblastoma specimens. To determine the role of the invasion-related genes, invasive activities were evaluated in vitro and in vivo. Fibroblast growth factor 13 (FGF13) was overexpressed in J3T-2 cells compared to J3T-1 cells, and in human glioma cell lines, human glioblastoma stem cells and human glioblastoma specimens, when compared to that of normal human astrocytes. Immunohistochemical staining and the RNA-seq (sequencing) data from the IVY Glioblastoma Atlas Project showed FGF13 expression in glioma cells in the invasive edges of tumor specimens. Also, the intracellular distribution was mainly in the cytoplasm of tumor cells and colocalized with tubulin. Overexpression of FGF13 stabilized tubulin dynamics in vitro and knockdown of FGF13 decreased glioma invasion both in vitro and in vivo and prolonged overall survival of several xenograft models. FGF13 was negatively regulated by hypoxic condition. Silencing of FGF13 also decreased in vivo bevacizumab-induced glioma invasion. In conclusion, FGF13 regulated glioma cell invasion and bevacizumab-induced glioma invasion, and could be a novel target for glioma treatment.
Collapse
Affiliation(s)
- Y Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - T Ichikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - K Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - S Inoue
- Department of Neurosurgery, Okayama City Hospital, Okayama, Japan
| | - J Ishida
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - T Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - T Shimizu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Y Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Y Hattori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - A Uneda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Y Matsumoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - H Michiue
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - I Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
41
|
Roles of Na +, Ca 2+, and K + channels in the generation of repetitive firing and rhythmic bursting in adrenal chromaffin cells. Pflugers Arch 2017; 470:39-52. [PMID: 28776261 DOI: 10.1007/s00424-017-2048-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/23/2017] [Indexed: 12/30/2022]
Abstract
Adrenal chromaffin cells (CCs) are the main source of circulating catecholamines (CAs) that regulate the body response to stress. Release of CAs is controlled neurogenically by the activity of preganglionic sympathetic neurons through trains of action potentials (APs). APs in CCs are generated by robust depolarization following the activation of nicotinic and muscarinic receptors that are highly expressed in CCs. Bovine, rat, mouse, and human CCs also express a composite array of Na+, K+, and Ca2+ channels that regulate the resting potential, shape the APs, and set the frequency of AP trains. AP trains of increasing frequency induce enhanced release of CAs. If the primary role of CCs is simply to relay preganglionic nerve commands to CA secretion, why should they express such a diverse set of ion channels? An answer to this comes from recent observations that, like in neurons, CCs undergo complex firing patterns of APs suggesting the existence of an intrinsic CC excitability (non-neurogenically controlled). Recent work has shown that CCs undergo occasional or persistent burst firing elicited by altered physiological conditions or deletion of pore-regulating auxiliary subunits. In this review, we aim to give a rationale to the role of the many ion channel types regulating CC excitability. We will first describe their functional properties and then analyze how they contribute to pacemaking, AP shape, and burst waveforms. We will also furnish clear indications on missing ion conductances that may be involved in pacemaking and highlight the contribution of the crucial channels involved in burst firing.
Collapse
|
42
|
Di Re J, Wadsworth PA, Laezza F. Intracellular Fibroblast Growth Factor 14: Emerging Risk Factor for Brain Disorders. Front Cell Neurosci 2017; 11:103. [PMID: 28469558 PMCID: PMC5396478 DOI: 10.3389/fncel.2017.00103] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/28/2017] [Indexed: 01/31/2023] Open
Abstract
The finely tuned regulation of neuronal firing relies on the integrity of ion channel macromolecular complexes. Minimal disturbances of these tightly regulated networks can lead to persistent maladaptive plasticity of brain circuitry. The intracellular fibroblast growth factor 14 (FGF14) belongs to the nexus of proteins interacting with voltage-gated Na+ (Nav) channels at the axonal initial segment. Through isoform-specific interactions with the intracellular C-terminal tail of neuronal Nav channels (Nav1.1, Nav1.2, Nav1.6), FGF14 controls channel gating, axonal targeting and phosphorylation in neurons effecting excitability. FGF14 has been also involved in synaptic transmission, plasticity and neurogenesis in the cortico-mesolimbic circuit with cognitive and affective behavioral outcomes. In translational studies, interest in FGF14 continues to rise with a growing list of associative links to diseases of the cognitive and affective domains such as neurodegeneration, depression, anxiety, addictive behaviors and recently schizophrenia, suggesting its role as a converging node in the etiology of complex brain disorders. Yet, a full understanding of FGF14 function in neurons is far from being complete and likely to involve other functions unrelated to the direct regulation of Nav channels. The goal of this Mini Review article is to provide a summary of studies on the emerging role of FGF14 in complex brain disorders.
Collapse
Affiliation(s)
- Jessica Di Re
- Neuroscience Graduate Program, University of Texas Medical BranchGalveston, TX, USA.,Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA
| | - Paul A Wadsworth
- Biochemistry and Molecular Biology Graduate Program, The University of Texas Medical BranchGalveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA.,Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical BranchGalveston, TX, USA.,Center for Addiction Research, The University of Texas Medical BranchGalveston, TX, USA
| |
Collapse
|
43
|
Inserra MC, Israel MR, Caldwell A, Castro J, Deuis JR, Harrington AM, Keramidas A, Garcia-Caraballo S, Maddern J, Erickson A, Grundy L, Rychkov GY, Zimmermann K, Lewis RJ, Brierley SM, Vetter I. Multiple sodium channel isoforms mediate the pathological effects of Pacific ciguatoxin-1. Sci Rep 2017; 7:42810. [PMID: 28225079 PMCID: PMC5320492 DOI: 10.1038/srep42810] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/13/2017] [Indexed: 01/04/2023] Open
Abstract
Human intoxication with the seafood poison ciguatoxin, a dinoflagellate polyether that activates voltage-gated sodium channels (NaV), causes ciguatera, a disease characterised by gastrointestinal and neurological disturbances. We assessed the activity of the most potent congener, Pacific ciguatoxin-1 (P-CTX-1), on NaV1.1–1.9 using imaging and electrophysiological approaches. Although P-CTX-1 is essentially a non-selective NaV toxin and shifted the voltage-dependence of activation to more hyperpolarising potentials at all NaV subtypes, an increase in the inactivation time constant was observed only at NaV1.8, while the slope factor of the conductance-voltage curves was significantly increased for NaV1.7 and peak current was significantly increased for NaV1.6. Accordingly, P-CTX-1-induced visceral and cutaneous pain behaviours were significantly decreased after pharmacological inhibition of NaV1.8 and the tetrodotoxin-sensitive isoforms NaV1.7 and NaV1.6, respectively. The contribution of these isoforms to excitability of peripheral C- and A-fibre sensory neurons, confirmed using murine skin and visceral single-fibre recordings, reflects the expression pattern of NaV isoforms in peripheral sensory neurons and their contribution to membrane depolarisation, action potential initiation and propagation.
Collapse
Affiliation(s)
- Marco C Inserra
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, Queensland 4072, Australia
| | - Mathilde R Israel
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, Queensland 4072, Australia
| | - Ashlee Caldwell
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Joel Castro
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Jennifer R Deuis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, Queensland 4072, Australia
| | - Andrea M Harrington
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Angelo Keramidas
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Jessica Maddern
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Andelain Erickson
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Luke Grundy
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Grigori Y Rychkov
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Katharina Zimmermann
- Klinik für Anästhesiologie am Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Richard J Lewis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, Queensland 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Group, South Australian Health and Medical Research Institute (SAHMRI), School of Medicine, Flinders University, Adelaide, South Australia 5000, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, Queensland 4072, Australia.,School of Pharmacy, The University of Queensland, 20 Cornwall St, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
44
|
FGF13 Selectively Regulates Heat Nociception by Interacting with Nav1.7. Neuron 2017; 93:806-821.e9. [DOI: 10.1016/j.neuron.2017.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/15/2016] [Accepted: 01/04/2017] [Indexed: 12/19/2022]
|
45
|
Barbosa C, Xiao Y, Johnson AJ, Xie W, Strong JA, Zhang JM, Cummins TR. FHF2 isoforms differentially regulate Nav1.6-mediated resurgent sodium currents in dorsal root ganglion neurons. Pflugers Arch 2016; 469:195-212. [PMID: 27999940 DOI: 10.1007/s00424-016-1911-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/19/2016] [Accepted: 11/20/2016] [Indexed: 10/20/2022]
Abstract
Nav1.6 and Nav1.6-mediated resurgent currents have been implicated in several pain pathologies. However, our knowledge of how fast resurgent currents are modulated in neurons is limited. Our study explored the potential regulation of Nav1.6-mediated resurgent currents by isoforms of fibroblast growth factor homologous factor 2 (FHF2) in an effort to address the gap in our knowledge. FHF2 isoforms colocalize with Nav1.6 in peripheral sensory neurons. Cell line studies suggest that these proteins differentially regulate inactivation. In particular, FHF2A mediates long-term inactivation, a mechanism proposed to compete with the open-channel blocker mechanism that mediates resurgent currents. On the other hand, FHF2B lacks the ability to mediate long-term inactivation and may delay inactivation favoring open-channel block. Based on these observations, we hypothesized that FHF2A limits resurgent currents, whereas FHF2B enhances resurgent currents. Overall, our results suggest that FHF2A negatively regulates fast resurgent current by enhancing long-term inactivation and delaying recovery. In contrast, FHF2B positively regulated resurgent current and did not alter long-term inactivation. Chimeric constructs of FHF2A and Navβ4 (likely the endogenous open channel blocker in sensory neurons) exhibited differential effects on resurgent currents, suggesting that specific regions within FHF2A and Navβ4 have important regulatory functions. Our data also indicate that FHFAs and FHF2B isoform expression are differentially regulated in a radicular pain model and that associated neuronal hyperexcitability is substantially attenuated by a FHFA peptide. As such, these findings suggest that FHF2A and FHF2B regulate resurgent current in sensory neurons and may contribute to hyperexcitability associated with some pain pathologies.
Collapse
Affiliation(s)
- Cindy Barbosa
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yucheng Xiao
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Johnson
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wenrui Xie
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA
| | - Judith A Strong
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA
| | - Jun-Ming Zhang
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA
| | - Theodore R Cummins
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
46
|
Dover K, Marra C, Solinas S, Popovic M, Subramaniyam S, Zecevic D, D'Angelo E, Goldfarb M. FHF-independent conduction of action potentials along the leak-resistant cerebellar granule cell axon. Nat Commun 2016; 7:12895. [PMID: 27666389 PMCID: PMC5052690 DOI: 10.1038/ncomms12895] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 08/11/2016] [Indexed: 12/31/2022] Open
Abstract
Neurons in vertebrate central nervous systems initiate and conduct sodium action potentials in distinct subcellular compartments that differ architecturally and electrically. Here, we report several unanticipated passive and active properties of the cerebellar granule cell's unmyelinated axon. Whereas spike initiation at the axon initial segment relies on sodium channel (Nav)-associated fibroblast growth factor homologous factor (FHF) proteins to delay Nav inactivation, distal axonal Navs show little FHF association or FHF requirement for high-frequency transmission, velocity and waveforms of conducting action potentials. In addition, leak conductance density along the distal axon is estimated as <1% that of somatodendritic membrane. The faster inactivation rate of FHF-free Navs together with very low axonal leak conductance serves to minimize ionic fluxes and energetic demand during repetitive spike conduction and at rest. The absence of FHFs from Navs at nodes of Ranvier in the central nervous system suggests a similar mechanism of current flux minimization along myelinated axons. FHFs are known to regulate voltage-gated sodium channels (NaVs). Here, the authors compare the role of FHFs in cerebellar granule cell propagation, and find NaVs in the distal axon function independently of FHFs, allowing for faster inactivation rates and reducing energy demands during repetitive spiking.
Collapse
Affiliation(s)
- Katarzyna Dover
- Department of Biological Sciences, Hunter College of City University, 695 Park Avenue, New York, New York 10065, USA.,Graduate Center of City University, Molecular, Cellular and Developmental Biology Subprogram, 365 Fifth Avenue, New York, New York 10016, USA
| | - Christopher Marra
- Department of Biological Sciences, Hunter College of City University, 695 Park Avenue, New York, New York 10065, USA.,Graduate Center of City University, Neuroscience Subprogram, 365 Fifth Avenue, New York, New York 10016, USA
| | - Sergio Solinas
- Brain Connectivity Center, C. Mondino National Neurological Institute, Via Forlanini 6, Pavia 27100, Italy
| | - Marko Popovic
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Sathyaa Subramaniyam
- Department of Brain and Behavioral Science, University of Pavia, Via Forlanini 6, Pavia 27100, Italy
| | - Dejan Zecevic
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Egidio D'Angelo
- Brain Connectivity Center, C. Mondino National Neurological Institute, Via Forlanini 6, Pavia 27100, Italy.,Department of Brain and Behavioral Science, University of Pavia, Via Forlanini 6, Pavia 27100, Italy
| | - Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, 695 Park Avenue, New York, New York 10065, USA.,Graduate Center of City University, Neuroscience Subprogram, 365 Fifth Avenue, New York, New York 10016, USA
| |
Collapse
|
47
|
Tanaka BS, Zhao P, Dib-Hajj FB, Morisset V, Tate S, Waxman SG, Dib-Hajj SD. A gain-of-function mutation in Nav1.6 in a case of trigeminal neuralgia. Mol Med 2016; 22:338-348. [PMID: 27496104 DOI: 10.2119/molmed.2016.00131] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/18/2016] [Indexed: 01/29/2023] Open
Abstract
Idiopathic trigeminal neuralgia (TN) is a debilitating pain disorder characterized by episodic unilateral facial pain along the territory of branches of the trigeminal nerve. Human painful disorders, but not TN, have been linked to gain-of-function mutations in peripheral voltage-gated sodium channels (NaV1.7, NaV1.8 and NaV1.9). Gain-of-function mutations in NaV1.6, which is expressed in myelinated and unmyelinated CNS and peripheral nervous system neurons and supports neuronal high-frequency firing, have been linked to epilepsy but not to pain. Here, we describe an individual who presented with evoked and spontaneous paroxysmal unilateral facial pain, and carried a diagnosis of TN. Magnetic resonance imaging showed unilateral neurovascular compression, consistent with pain in areas innervated by the second branch of the trigeminal nerve. Genetic analysis as part of a phase 2 clinical study in patients with TN conducted by Convergence Pharmaceuticals Ltd revealed a previously undescribed de novo missense mutation in NaV1.6 (c.A406G; p.Met136Val). Whole-cell voltage-clamp recordings show that the Met136Val mutation significantly increases peak current density (1.5-fold) and resurgent current (1.6-fold) without altering gating properties. Current-clamp studies in trigeminal ganglion (TRG) neurons showed that Met136Val increased the fraction of high-firing neurons, lowered the current threshold and increased the frequency of evoked action potentials in response to graded stimuli. Our results demonstrate a novel NaV1.6 mutation in TN, and show that this mutation potentiates transient and resurgent sodium currents and leads to increased excitability in TRG neurons. We suggest that this gain-of-function NaV1.6 mutation may exacerbate the pathophysiology of vascular compression and contribute to TN.
Collapse
Affiliation(s)
- Brian S Tanaka
- Department of Neurology.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT 06516
| | - Peng Zhao
- Department of Neurology.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT 06516
| | - Fadia B Dib-Hajj
- Department of Neurology.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT 06516
| | - Valerie Morisset
- Convergence Pharmaceuticals Ltd, a Biogen company, Cambridge, United Kingdom
| | - Simon Tate
- Convergence Pharmaceuticals Ltd, a Biogen company, Cambridge, United Kingdom
| | - Stephen G Waxman
- Department of Neurology.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT 06516
| | - Sulayman D Dib-Hajj
- Department of Neurology.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT 06516
| |
Collapse
|
48
|
Affiliation(s)
- Sulayman D Dib-Hajj
- a Center for Neuroscience & Regeneration Research, Yale Medical School and Veterans Affairs Hospital , West Haven , CT , USA
| |
Collapse
|
49
|
Pablo JL, Wang C, Presby MM, Pitt GS. Polarized localization of voltage-gated Na+ channels is regulated by concerted FGF13 and FGF14 action. Proc Natl Acad Sci U S A 2016; 113:E2665-74. [PMID: 27044086 PMCID: PMC4868475 DOI: 10.1073/pnas.1521194113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clustering of voltage-gated sodium channels (VGSCs) within the neuronal axon initial segment (AIS) is critical for efficient action potential initiation. Although initially inserted into both somatodendritic and axonal membranes, VGSCs are concentrated within the axon through mechanisms that include preferential axonal targeting and selective somatodendritic endocytosis. How the endocytic machinery specifically targets somatic VGSCs is unknown. Here, using knockdown strategies, we show that noncanonical FGF13 binds directly to VGSCs in hippocampal neurons to limit their somatodendritic surface expression, although exerting little effect on VGSCs within the AIS. In contrast, homologous FGF14, which is highly concentrated in the proximal axon, binds directly to VGSCs to promote their axonal localization. Single-point mutations in FGF13 or FGF14 abrogating VGSC interaction in vitro cannot support these specific functions in neurons. Thus, our data show how the concerted actions of FGF13 and FGF14 regulate the polarized localization of VGSCs that supports efficient action potential initiation.
Collapse
Affiliation(s)
- Juan Lorenzo Pablo
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710; Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710
| | - Chaojian Wang
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710; Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Matthew M Presby
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710; Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Geoffrey S Pitt
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710; Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710; Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
50
|
Siekierska A, Isrie M, Liu Y, Scheldeman C, Vanthillo N, Lagae L, de Witte PAM, Van Esch H, Goldfarb M, Buyse GM. Gain-of-function FHF1 mutation causes early-onset epileptic encephalopathy with cerebellar atrophy. Neurology 2016; 86:2162-70. [PMID: 27164707 DOI: 10.1212/wnl.0000000000002752] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/12/2016] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE Voltage-gated sodium channel (Nav)-encoding genes are among early-onset epileptic encephalopathies (EOEE) targets, suggesting that other genes encoding Nav-binding proteins, such as fibroblast growth factor homologous factors (FHFs), may also play roles in these disorders. METHODS To identify additional genes for EOEE, we performed whole-exome sequencing in a family quintet with 2 siblings with a lethal disease characterized by EOEE and cerebellar atrophy. The pathogenic nature and functional consequences of the identified sequence alteration were determined by electrophysiologic studies in vitro and in vivo. RESULTS A de novo heterozygous missense mutation was identified in the FHF1 gene (FHF1AR114H, FHF1BR52H) in the 2 affected siblings. The mutant FHF1 proteins had a strong gain-of-function phenotype in transfected Neuro2A cells, enhancing the depolarizing shifts in Nav1.6 voltage-dependent fast inactivation, predicting increased neuronal excitability. Surprisingly, the gain-of-function effect is predicted to result from weaker interaction of mutant FHF1 with the Nav cytoplasmic tail. Transgenic overexpression of mutant FHF1B in zebrafish larvae enhanced epileptiform discharges, demonstrating the epileptic potential of this FHF1 mutation in the affected children. CONCLUSIONS Our data demonstrate that gain-of-function FHF mutations can cause neurologic disorder, and expand the repertoire of genetic causes (FHF1) and mechanisms (altered Nav gating) underlying EOEE and cerebellar atrophy.
Collapse
Affiliation(s)
- Aleksandra Siekierska
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Mala Isrie
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Yue Liu
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Chloë Scheldeman
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Niels Vanthillo
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Lieven Lagae
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Peter A M de Witte
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Hilde Van Esch
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Mitchell Goldfarb
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY
| | - Gunnar M Buyse
- From the Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences (A.S., C.S., N.V., P.A.M.d.W.), and Laboratory for the Genetics of Cognition (M.I.), University of Leuven; Center for Human Genetics (M.I., H.V.E.) and Child Neurology (L.L., G.M.B.), University Hospitals Leuven; Department of Biological Sciences (Y.L., M.G.), Hunter College of City University, New York; and Graduate Program in Biology/Neuroscience at City University (Y.L.), New York, NY.
| |
Collapse
|