Skip to main content

Main menu

  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE

User menu

  • Log out
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Neuroscience
  • Log out
  • Log in
  • My Cart
Journal of Neuroscience

Advanced Search

Submit a Manuscript
  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE
PreviousNext
Featured ArticleResearch Articles, Cellular/Molecular

Cofilin Inhibition Ameliorates PIEZO2 and AMPA Dysfunction in a Mouse Model of Angelman Syndrome

Luis O. Romero, Manisha Bade, Elisa Carrillo, Sonia Paz-López, Syed A. M. Hasan, William James Antonisamy, Vasanthi Jayaraman, Zahoor A. Shah, Valeria Vásquez and Julio F. Cordero-Morales
Journal of Neuroscience 5 November 2025, 45 (45) e0965252025; https://doi.org/10.1523/JNEUROSCI.0965-25.2025
Luis O. Romero
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Luis O. Romero
Manisha Bade
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
2The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Manisha Bade
Elisa Carrillo
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Elisa Carrillo
Sonia Paz-López
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Sonia Paz-López
Syed A. M. Hasan
3Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
William James Antonisamy
3Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Vasanthi Jayaraman
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Vasanthi Jayaraman
Zahoor A. Shah
3Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Zahoor A. Shah
Valeria Vásquez
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Valeria Vásquez
Julio F. Cordero-Morales
1Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Julio F. Cordero-Morales
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • Peer Review
  • PDF
Loading

Abstract

Angelman syndrome (AS) is a neurogenetic disorder characterized by motor coordination and cognitive deficits. In AS, hippocampal neurons show reduced filamentous (F-)actin, a decrease we also reported in dorsal root ganglia (DRG) neurons, along with impaired mechanosensitive ion channel activity. Currently, there are no pharmacological targets to prevent the decrease of F-actin in AS. Here, we utilize a first-in-class selective cofilin inhibitor (SZ-3) to restore PIEZO2 function in DRG neurons and glutamate-evoked currents in hippocampal neurons from AS mice. Using atomic force microscopy, we demonstrate that inhibiting cofilin, an actin-severing protein, with SZ-3 increases cellular stiffness by stabilizing the actin cytoskeleton. Furthermore, systemic administration of SZ-3 in male and female AS mice enhances their performance in the rotarod and T-maze tests. These findings support that cytoskeletal dysregulation contributes to impaired ion channel function and behavioral deficits and that actin-binding proteins could serve as a target for enhancing motor coordination and spatial learning in AS.

  • AMPA receptor
  • Angelman syndrome
  • cofilin
  • hippocampal neurons
  • PIEZO2
  • sensory neurons

Significance Statement

Angelman syndrome (AS) is a severe neurogenetic disorder characterized by significant motor and cognitive impairments; however, effective treatments remain elusive. Recent evidence implicates deficits in the mechanosensitive PIEZO2 channel and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor function, as well as cytoskeletal abnormalities in AS pathology. Our study identifies cofilin, an actin-binding protein, as a regulator of ion channel function. We demonstrate that pharmacological inhibition of cofilin restores PIEZO2 channel and AMPA receptor activities, enhances neuronal excitability, and improves motor coordination and learning in a mouse model of AS. These findings reveal a novel mechanism by which actin dynamics influence sensory and cognitive function.

Introduction

UBE3A is an E3 ubiquitin ligase that transfers a ubiquitin molecule from E2 proteins to substrates, tagging them for proteasomal degradation (Scheffner et al., 1993; Lopez et al., 2018). Loss-of-function (LOF) mutations or the loss of UBE3A expression result in Angelman syndrome (AS; Knoll et al., 1989; Kishino et al., 1997; Clayton-Smith and Laan, 2003). AS is a neurogenetic disorder featuring cognitive dysfunction, impaired motor coordination, balance deficits, gastrointestinal dysfunction, and seizures (Tan et al., 2011; Larson et al., 2015; Buiting et al., 2016; Wheeler et al., 2017; Duis et al., 2022). UBE3A is genetically imprinted (i.e., a process that leads to monoallelic gene expression) in most neurons of the central nervous system (CNS) and in large-diameter mechanoreceptor neurons of the dorsal root ganglia (DRG; Lopez et al., 2017; McCoy et al., 2017; Tucci et al., 2019). Thus, loss of the maternal allele yields UBE3A-deficient neurons, leading to AS (Buiting et al., 2016).

Proprioceptive behaviors are among the affected sensory processing patterns observed in individuals with AS (Walz and Baranek, 2006). PIEZO2 is a mechanosensitive ion channel expressed in DRG neurons that innervate the skin and muscle spindles, where it mediates touch and proprioception (Coste et al., 2010; Ranade et al., 2014; Woo et al., 2014, 2015; Chesler et al., 2016; Szczot et al., 2021; Nagel and Chesler, 2022). Humans with PIEZO2 LOF and individuals with AS exhibit similar proprioceptive deficits, including impaired balance and unsteady gait (Larson et al., 2015; Chesler et al., 2016; Wheeler et al., 2017). Recently, we proposed a model in which the loss of UBE3A expression, in DRG neurons from an AS mouse model, increases cofilin, decreases filamentous (F-)actin, and consequently downregulates PIEZO2 membrane expression and function (Romero et al., 2023). Importantly, PIEZO2 activity is modulated by the cytoskeleton (Romero et al., 2020; Wang et al., 2022). Hence, interventions aimed at stabilizing F-actin may enhance PIEZO2 activity and improve neuronal function.

Individuals with AS and the corresponding mouse models show impaired synaptic plasticity, which is thought to contribute to the cognitive dysfunction underlying their abnormal spatial and context-dependent learning (Jiang et al., 1998; Miura et al., 2002; Fink et al., 2017). The α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors are glutamate-gated cation channels that mediate fast excitatory transmission in the CNS (Henley and Wilkinson, 2013, 2016; Diering and Huganir, 2018), and a decrease in their activity has been associated with cognitive decline (Chang et al., 2006; Henley and Wilkinson, 2013; Ding et al., 2016). Moreover, the loss of Ube3a in neurons results in decreased activity of AMPA receptors (Greer et al., 2010). Taken together, dysregulation of AMPA receptors may contribute to the cognitive deficits observed in AS, underscoring the potential for novel strategies aimed at increasing AMPA receptor activity.

Actin regulates membrane protein trafficking (Cingolani and Goda, 2008; Zech et al., 2012; Sasaki et al., 2014; Aiken and Holzbaur, 2021). Indeed, disrupting F-actin with latrunculin A reduces PIEZO2 membrane expression and function (Romero et al., 2023). Cofilin is an actin-binding protein that promotes the disassembly of F-actin by binding to and severing it into shorter fragments (Bamburg, 1999; Maciver and Hussey, 2002; Huang et al., 2006; Wioland et al., 2017). We recently developed the first-in-class selective cofilin inhibitor (SZ-3) that reduces cofilin-mediated F-actin depolymerization both in vitro and in vivo (Alaqel et al., 2022; Bahader et al., 2023; Almarghalani et al., 2024). SZ-3 treatment improved neurological and cognitive functions following intracerebral hemorrhage and traumatic brain injury in mice (Bahader et al., 2023; Almarghalani et al., 2024). Moreover, we have shown that SZ-3 can protect microglial and neuroblastoma cells from thrombin-induced apoptosis in vitro (Alaqel et al., 2022). Although the loss of the maternally inherited UBE3A gene disrupts several proteins, our study focuses specifically on PIEZO2 and AMPA receptors. We identify that cofilin activity regulates both channels in the context of AS. Pharmacological inhibition of cofilin with SZ-3 enhances neuronal function and improves motor and cognitive performance.

Material and methods

Animals

Mice procedures described below were reviewed and approved by the University of Texas Health Science Center at Houston (UTHealth) Animal Welfare Committee (UTHealth AWC protocol number, AWC-23-0093) and the Center for Laboratory Animal Medicine and Care and by the Institutional Animal Care and Utilization Committee at the University of Toledo Health Science Campus (Ethics Reference Number 400095-01-UT). All methods were carried out according to approved guidelines. Adult (2- to 6-month-old) mice were housed with a 12 h light/dark cycle at 21°C with 40–60% humidity and with food and water ad libitum. We bred mice from our in-house colony, and their genotype was confirmed using previously published protocols (Jiang et al., 1998; Judson et al., 2016). We used a two-step breeding scheme. Briefly, we first crossed female WT with male Ube3am+/p− to generate heterozygous offspring, in which half of the progeny were Ube3am+/p−, with no AS phenotype. Ube3am+/p− heterozygous females were crossed with WT males to generate Ube3am−/p+ mice, in which half of the progeny constitute AS mice displaying the corresponding phenotype. After confirming the genotype, the progenies were used for experiments. We also used the WT mouse strain (C57BL/6J; Stock Number 000664) and Ube3am–/p+ mice obtained from The Jackson Laboratory, strain C57BL/6 Ube3atm1Alb (B6 AS; Stock Number 016590).

Mouse DRG neurons

Primary cultures of mouse DRG neurons were obtained from 8–12-week-old male Ube3am–/p+ (maternal transmission) mice. Mice were anesthetized with isoflurane and killed by cervical dislocation. DRGs were dissected and kept on ice in 1× Hank’s balanced salt solution (HBSS without CaCl2 and MgCl2). Next, DRGs were incubated with 1 mg/ml collagenase B (Sigma-Aldrich) in HBSS at 37°C and 5% CO2 and then, after 1 h, were dissociated in a medium without serum. The cell suspension solution was centrifuged for 8 min at 62 × g. The pellet was resuspended in Dulbecco’s modified Eagle medium (DMEM; Invitrogen) complete media containing 1% penicillin–streptomycin (Invitrogen), 1% MEM vitamin solution (Invitrogen), 1% l-glutamine (Invitrogen), and 10% horse serum (Invitrogen). Cells were plated on poly-l-lysine (Sigma-Aldrich)-treated glass coverslips in 24-well plates. All mouse DRG neurons were kept at 37°C, with 95% relative humidity and 5% CO2. Cells were used in experiments after 24–48 h.

siRNA-mediated knockdown

Primary cultures of mouse DRG neurons were transfected with Lipofectamine RNAiMAX Transfection Reagent (Thermo Fisher Scientific), according to the manufacturer’s protocols. The siRNA concentration was 60 nM for Cofilin (AA959946, Horizon Discovery Ltd.) or the silencer negative control. The transfections were done with antibiotic-free media. After 6 h of transfection, the medium was replaced with fresh media containing antibiotics. All cells were used 48 h after transfection. For electrophysiology experiments, cells were also cotransfected with siGLO Green Transfection Indicator (Dharmacon).

Cell culture

Human Merkel cell carcinoma cell line (MCC13; Cell Bank Australia reference number, CBA1338) was obtained from Sigma-Aldrich and cultured according to the manufacturer’s protocol. MCC13 cells were cultured in RPMI1640 (with 2 mM l-glutamine and 25 mM HEPES; Sigma-Aldrich), 5% penicillin–streptomycin, and 10% FBS. Human embryonic kidney cells (HEK293) obtained from the American Type Culture Collection were cultured in DMEM, supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin. All cultured cells were maintained at 37°C, with 95% relative humidity and 5% CO2. Cells were incubated in media supplemented with 10 µM SZ-3 for 18 h before G-/F-actin quantification, atomic force microscopy (AFM), and electrophysiology experiments.

Cell transfections

For electrophysiology, we used Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer’s instructions. MCC13 cells were grown in six-well plates to 90% confluency and cotransfected with 1.2 µg/ml of Z-lock cofilin (Addgene #141137) and 50 ng/ml GFP-pMO (a pcDNA3.1-based vector with the 5′ and 3′ untranslated regions of the beta-globin gene). HEK 293 cells were grown in six-well plates to 70% confluency and cotransfected with 1 µg/ml AMPA and NMDA receptors, 50 ng/ml rTRPV1-pMO, 500 ng/ml hTRPA1-pMO, or 1 µg/ml rASIC1and 20–50 ng/ml GFP-pMO.

Protein expression determination

The cytoskeletal fraction of SZ-3- (10 µM, 18 h) and DMSO-treated MCC13 cells was extracted with the globular (G-)actin/ F-actin in vivo assay kit (Cytoskeleton) to determine the G- and F-actin forms according to the manufacturer's instructions. The two fractions of each sample were loaded in equivalent volumes in Mini-PROTEAN TGX Stain-free Precast Gels (Bio-Rad Laboratories) and dry transferred to an Immun-Blot PVDF Membrane (Bio-Rad Laboratories). Mouse monoclonal anti-actin (1:1,000; Cytoskeleton; catalog #AAN02-S; RRID, AB_2884962) and goat polyclonal anti-mouse IgG H&L (1:20,000; Abcam catalog #ab205719; RRID, AB_2755049) antibodies were used for Western blot. Membranes were developed with Clarity Western ECL Substrate and imaged in a ChemiDoc Touch Imaging System (Bio-Rad Laboratories) for chemiluminescence. Western blot images were then quantified using the Image Lab software (v6.1.0; Bio-Rad Laboratories) by comparing the globular fraction's signal with each sample's filamentous fraction (G/F ratio).

Human iPSC-derived sensory neurons

The human-induced pluripotent stem cell (hiPSC) line was manufactured and characterized at Anatomic Incorporated facilities with informed consent, using proprietary technologies. hiPSCs were differentiated into sensory neurons using a commercially available kit, Senso-DM, according to the manufacturer’s instructions (Anatomic Incorporated, #7007). Sensory neurons were maintained in culture at 37°C with 5% CO2 on glass coverslips coated with poly-l-lysine and Matrix 3 (Anatomic Incorporated, #M8003) in Senso-MM media (Anatomic Incorporated, #7008). hiPSC-derived sensory neurons were incubated in culture for 14 d before recording. Neurons were incubated in media supplemented with 5 µM SZ-3 for 18 h before electrophysiology experiments.

Hippocampal neurons

Hippocampal neurons were dissected from early postnatal day (P1–P4) Ube3am–/p+ mice, either sex (Carrillo et al., 2020, 2021). The mice were decapitated, the skin and skull were removed, and the brain was placed in 4–5 ml ice-cold dissection solution (Earle's buffered salt solution, Invitrogen, 10 mm HEPES). Under a dissecting microscope, the hippocampus was removed and transferred to a solution containing 10 U/ml papain (Worthington), 20 µl DNase I (New England Biolabs, 2,000 U/mg), and 50 µM APV (Abcam) and incubated in a 37°C water bath for 30 min. Every 10 min, the tissue was pipetted to dissociate into individual cells. The activity of papain was stopped by the addition of 100% inactivated FBS for 2 min. The tissue was centrifuged and transferred to a dissociation solution containing B27/Neurobasal culture medium (Invitrogen), supplemented with 2 mM l-glutamine, 0.2% penicillin–streptomycin, and 20 mM glucose. The cell suspension was diluted with incubated feeding medium (B27/Neurobasal culture medium, supplemented with 2 mM l-glutamine, and 0.2% penicillin–streptomycin), plated on poly-l-lysine and laminin (Cell Applications)-coated glass coverslips, and incubated in 5% CO2 at 37 °C. Neurons were used 24 h after plating. Those used in experiments with SZ-3 (5 µM) compound were incubated for 3 h before recording.

Electrophysiology

Patch-clamp recordings were performed on cells plated on glass coverslips. For whole-cell recordings of mechano-activated currents in the voltage-clamp mode, the bath solution contained (in mM) 140 NaCl, 6 KCl, 2 CaCl2, 1 MgCl2, 10 glucose, and 10 HEPES, pH 7.4, while the pipette solution contained (in mM) 140 CsCl, 5 EGTA, 1 CaCl2, 1 MgCl2, and 10 HEPES, pH 7.2. For current-clamp recordings of action potentials elicited by mechanical stimulation, the bath solution contained (in mM) 140 NaCl, 6 KCl, 2 CaCl2, 1 MgCl2, 10 glucose, and 10 HEPES, pH 7.4, while the pipette solution contained (in mM) 140 KCl, 6 NaCl, 2 CaCl2, 1 MgCl2, 10 glucose, and 10 HEPES, pH 7.4. Mechanical stimulation was performed using the voltage-clamp (constant −60 mV) or current-clamp configuration. Mechano-activated currents were sampled at 100 kHz and low-pass filtered at 10 kHz using a MultiClamp 700 B amplifier and Clampex (v10.4.2.0; Molecular Devices). Leak currents, before mechanical stimulation, were subtracted offline from the current traces, and data were digitally filtered at 2 kHz with ClampFit (v10.4.2.0; Molecular Devices). Recordings with leak currents >200 pA, with access resistance >10 MΩ, and cells with giga-seals that did not withstand at least five consecutive steps of mechanical stimulation were excluded from analyses.

For AMPA receptor recordings of hippocampal neurons, measurements were performed with borosilicate pipettes with a resistance between 8 and 15 mΩ, filled with internal solution as follows (in mM): 135 CsF, 33 CsCl, 2 MgCl2, 1 CaCl2, 11 EGTA, 10 HEPES, 0.4 GTP-Na, 4 ATP-Mg, and 5 phosphocreatine, pH 7.3. The bath solution contained 150 mM NaCl, 6 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 10 mM HEPES and 10 mM glucose, 50 mM APV, 10 µM bicuculline, and 1 mM TTX, pH 7.4. The external solutions were locally applied to neurons using a SF-77B perfusion Fast-Step (Warner Instruments). All hippocampal neuron recordings were performed at room temperature with a holding potential of −60 mV using an Axopatch 200B amplifier, acquired at 10 kHz using pCLAMP10.7 software, and filtered online at 5 kHz.

For NMDA receptor currents, in transfected HEK293 cells, the pipette solution was comprised of (in mM) 135 CsF, 33 CsOH, 2 MgCl2, 1 CaCl2, 11 EGTA, and 10 HEPES, adjusted to pH 7.4 with CsOH. The bath solution contains the following (in mM): 140 NaCl, 2.8 KCl, 1 CaCl2, and 10 HEPES, adjusted to pH 7.4 with NaOH. The 1 mM glutamate and 1 mM glycine were applied to lifted cells using a stepper motor system (SF-77B, Warner Instruments) with triple-barrel tubing. For ASICs currents, in transfected HEK293 cells, the pipette solution was comprised of (in mM) 135 CsF, 33 CsOH, 2 MgCl2, 1 CaCl2, 11 EGTA, and 10 HEPES, adjusted to pH 7.4 with CsOH. The bath solution contains the following (in mM): 150 NaCl, 20 HEPES, 1 CaCl2, and 1 MgCl2 adjusted to pH 7.4, 6.0, or 5.0. Recordings were performed using an Axopatch 200B amplifier at −60 mV hold potential, acquired at 10 kHz using pCLAMP10 software, and filtered online at 5 kHz.

For TRP channels, transfected HEK293 cells, the bath solution contained (in mM) 140 NaCl, 6 KCl, 1 MgCl2, 10 glucose, and 10 HEPES, pH 7.4, while the pipette solution contained (in mM) 140 CsCl, 5 EGTA, and 10 HEPES, pH 7.2. Recordings were sampled at 100 kHz and low-pass filtered at 10 kHz using a MultiClamp 700 B amplifier and Clampex.

Current densities were calculated by dividing the current value (in picoamperes) by the measured cell membrane capacitance (picofarads). Before recording, cells were incubated with SZ-3 (5 or 10 µM for 18 h) or a control solution (DMSO alone), as indicated in each subsection of the methods.

Mechanical stimulation

For indentation assays, DRG neurons, MCC13, and hiPSC-derived sensory neurons were mechanically stimulated with a heat-polished blunt glass pipette (3–4 µm) driven by a piezo servo controller (E625, Physik Instrumente). The blunt pipette was mounted on a micromanipulator at a ∼45° angle and positioned 3–4 µm above the cells. Displacement measurements were obtained with a square-pulse protocol consisting of 1 µm incremental indentation steps, each lasting 200 ms with a 2 ms ramp in 10 s intervals. The threshold of mechano-activated currents for each experiment was defined as the indentation step that evoked the first current deflection from the baseline. For current-clamp experiments, the mechanical threshold was defined as the indentation step that produced the first action potential. Only cells that did not detach throughout the stimulation protocols were included in the analyses. The piezo servo controller was automated, using a MultiClamp 700B amplifier, through Clampex (Molecular Devices).

AFM

Atomic force microscopy (AFM) experiments were carried out with a BioScope II Controller (Bruker), operated with the Research NanoScope software version 7.30 integrated to a Nikon TE2000-E inverted optical microscope (Nikon Instruments) to facilitate bright-field/fluorescence imaging. Bead sphere colloid cantilever probes (Novascan PT.GS) consisted of a 5-µm-diameter borosilicate glass particle attached to the edge of a silicon nitride V-shaped cantilever with a nominal spring constant of 0.24 N/m. Each cantilever is calibrated for its laser sensitivity using the thermal oscillation method before each experiment. Indentation curves were captured using a 4 µm ramp size, at a scan rate of 0.5 Hz, and a trigger threshold with a maximum load of 10 nN. Young's modulus values were calculated following the Hertz model (spherical indenter radius, 2.5 µm) with a Poisson's ratio of 0.5, using the NanoScope Analysis software version 3.0 (Copyright Bruker).

Intraperitoneal injections

Cofilin inhibitor was dissolved in a vehicle solution that consisted of 4.9% DMSO, 4.9% Tween 20, and 88.9% 2-hydroxypropyl-β-cyclodextrin, yielding a final concentration of 1.3%. Ube3am–/p+ mice between the ages of 3 and 4 months were administered SZ-3 or vehicle for 10 d, at the same time each day by a technician at the UT Health mouse facility. For T-maze behavioral experiments, mice received SZ-3 at a dose of 10 mg/kg. For rotarod and von Frey assays, a dose of 25 mg/kg was administered.

Behavior

For rotarod, mice were acclimated to the experimental room for 1 h before the test. Mice were trained for 3 d by placing them on a rod (SDI rotor-rod, San Diego Instruments) that rotated at a constant speed of 4 rpm for 2 min. On the test day, mice were placed on the rod, which accelerated from 4 to 40 rpm in 5 min, and their latency to fall was recorded. Three trials were taken of each mouse with a 15 min interval between each trial. The average of three trials was used for analysis. The baseline behavior was recorded before the mice were administered the drug or vehicle and after 10 d of intraperitoneal injections. For T-maze, testing was performed by placing a mouse on the lower end of the T-maze and allowing it to choose one side spontaneously. After the initial choice, they are closed off with an insert to let them explore the arm of their choice for 30 s. Afterward, they are put back in their home cages for ∼2–3 min. Meanwhile, the researcher wipes the maze down with a paper towel to avoid odor queues and clean feces or urine. Then, mice are placed back in the start arm of the T-maze and tasked to make another choice. Choosing the same arm of the maze was counted as a failed alternation. If they choose the opposite arm, this was counted as a successful alternation. Each mouse repeated this protocol four times. Each mouse was habituated to the maze for 5 min the day before testing. Testing was performed on Day 7 and Day 12. For von Frey, mice were acclimated to the behavioral testing room and individually placed in modular holding chambers constructed from 3-mm-thick PVC positioned atop a metal mesh platform. Following a 1 h habituation period, von Frey filaments were applied to the plantar surface of both hindpaws in an ascending order of force. Each filament was applied three times per paw. A positive response, defined as a clear withdrawal or licking of the paw, was recorded if it occurred in at least two of the three applications. If no positive response was elicited, the next filament of greater force was applied. The paw withdrawal threshold was defined as the minimal force that evoked a positive response in two out of three trials. Final threshold values were reported as the average of two independent measurements taken from both the left and right hindpaws.

Data analysis

Data were plotted using OriginPro (2018 v:b9.51.195; OriginLab) and Estimation Stats (Ho et al., 2019). The time constant of inactivation τ was obtained by fitting a single exponential function between the peak value of the current at the end of the stimulus as follows:f(t)=∑i=1nAi*e−tτi+C, where A is the amplitude, τ is the time constant, and C is the constant y-offset for each component i. All boxplots show mean (square), median (bisecting line), bounds of box (75th to 25th percentiles), and outlier range with 1.5 coefficient (whiskers). Statistical analyses were performed using the GraphPad InStat software (version 3.10; GraphPad Software) and Estimation Stats (Ho et al., 2019). We used the Kolmogorov and Smirnov method to determine data distribution, as well as the Bartlett’s test to determine differences between standard deviations. Individual tests are described in each of the figure legends.

Statistical analysis

No statistical method was used to predetermine the sample size, and no data were excluded from the analyses. The experiments were not randomized. For electrophysiology, the investigator was blind to genotype and treatment. For behavioral assays, the investigators were blind when possible. All attempts at replication were successful. Experiments were performed at least three times on different days with different/independent preparations.

Data availability

The source data underlying figures are provided as a Source Data file and deposited at https://doi.org/10.6084/m9.figshare.28908632.

Results

Genetic manipulation of cofilin modulates PIEZO2 function

We demonstrated that the loss of UBE3A expression leads to increased cofilin levels and decreased PIEZO2 currents (Romero et al., 2023). Here, we used mice carrying a LOF Ube3a mutation on the maternal allele in a C57BL/6 genetic background (Jiang et al., 1998). Heterozygous mice with a maternal deficiency (Ube3am–/p+) exhibit AS-associated phenotypes, including a lack of balance and coordination (Jiang et al., 1998; Sonzogni et al., 2018). We aimed to determine whether reducing cofilin expression through siRNA-mediated silencing in DRG neurons from Ube3am–/p+ mice could restore mechanocurrents (Fig. 1A). Our results showed that all mechanocurrents, including the rapidly inactivating currents associated with PIEZO2, were significantly enhanced after cofilin knockdown compared with scrambled siRNA controls (Fig. 1B,C). The distribution of DRG neurons exhibiting rapidly inactivating, intermediate, and slow mechanocurrents was similar in neurons treated with scrambled and cofilin siRNA (Fig. 1D).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Knocking down cofilin increases PIEZO2 function. A, Micrographs showing mouse Ube3am–/p+ DRG neurons (top) transfected with cofilin siRNA and the transfection marker siGLO green (bottom). Micrographs are representative of at least 10 independent preparations. Black and white arrows highlight a positively transfected neuron. B, Representative whole-cell patch–clamp recordings elicited by mechanical stimulation (−60 mV) of rapidly, intermediate, and slowly inactivating currents of Ube3am–/p+ DRG neurons transfected with scrambled or cofilin siRNAs. C, Current densities elicited by maximum displacement of Ube3am–/p+ DRG neurons, transfected with scrambled or cofilin siRNAs, classified by their time constant of inactivation. Error bars indicate mean ± SD. Kruskal–Wallis (H = 31.45; p = 0.00012) and Dunn’s multiple-comparison test. D, Proportions of Ube3am–/p+ DRG neurons, transfected with scrambled or cofilin siRNA, classified by their time constant of inactivation of mechano-activated currents elicited by maximum displacement. χ2 test (χ2 = 0.23; p = 0.89). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

Optogenetic activation of cofilin decreases PIEZO2 currents

To assess whether the effect of cofilin could also occur acutely, we utilized an optogenetic approach to rapidly modulate its actin-severing ability with light using the Z-lock cofilin construct (Stone et al., 2019). In dark conditions, the interaction of Zdk (a protein A fragment) with the LOV2 domain of Avena sativa phototropin 1, linked to the C- and N-termini of cofilin, occludes the active site of cofilin (Stone et al., 2019). Blue light irradiation (400–500 nm) disrupts this interaction, exposing cofilin’s active site (Fig. 2A, top panel). We measured mechanocurrents using the whole-cell patch–clamp configuration from MCC13 cells that were transiently transfected with Z-lock cofilin. Measurements were taken in the dark at 0, 5, and 15 min and after exposure to blue light for 5 and 15 min (450 nm; Fig. 2A, bottom panel). Importantly, we observed a time-dependent reduction in mechanocurrent amplitudes following the photoactivation of Z-lock cofilin (Fig. 2B,C). Control experiments in cells lacking Z-lock cofilin showed no effect from blue light exposure, confirming that illumination alone did not affect mechanoresponses (Fig. 2B,C). Our findings indicate that acute cofilin activation decreases mechanocurrents.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Cofilin photoactivation decreases mechanosensitivity in MCC13 cells. A, Top, diagram describing the photoswitchable Z-lock cofilin. The light oxygen voltage (LOV) domain and Zdk, a small protein that selectively binds LOV in the dark, are appended to cofilin, where they sterically block the active site. Made with BioRender.com. Bottom, Timeline depicting the experimental design for modulation of mechanoresponses by photoswitchable Z-lock cofilin. B, Representative whole-cell patch–clamp recordings elicited by mechanical stimulation (12 µm; −60 mV) of MCC13 cells untransfected or transfected with Z-lock cofilin and recorded in the dark or after exposure to blue light (450 nm) as described in A. C, Left, current changes elicited by 12 µm displacement of MCC13 cells transfected with Z-lock cofilin exposed to blue light. Data samples are paired. Repeated-measure ANOVA (F = 159.55; p = 1.33−11) with Tukey–Kramer multiple-comparison test. Middle, Current changes elicited by 12 µm displacement of MCC13 cells transfected with Z-lock cofilin in the dark. Right, Current changes elicited by 12 µm displacement of MCC13 cells untransfected, exposed to blue light. Data samples are paired. Repeated-measures ANOVA (F = 3.92; p = 0.029) with Tukey–Kramer multiple-comparison test. n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

A first-in-class cofilin inhibitor enhances PIEZO2 currents

Since we observed an inverse relationship between cofilin expression and PIEZO2 function (Figs. 1, 2), we hypothesized that cofilin inhibition could enhance mechanocurrents and compensate for the mechanosensory deficits caused by the loss of UBE3A expression in AS. Recently, we developed SZ-3, a first-in-class small–molecule cofilin inhibitor (Fig. 3A,B; Alaqel et al., 2022). MCC13 cells treated with SZ-3 displayed a reduced G-/F-actin ratio (i.e., more F-actin), as confirmed by Western blots (Fig. 3C,D). AFM further demonstrated that SZ-3 treatment increases the Young’s modulus (i.e., stiffness) of MCC13 cells compared with the control (DMSO; Fig. 3E). On the other hand, treatment of MCC13 with latrunculin A, which disrupts F-actin, resulted in a decrease in the Young’s modulus compared with the control (EtOH; Fig. 3F). The increased F-actin and Young’s modulus observed with SZ-3 treatment may enhance mechanosensitive channel function. Indeed, MCC13 cells challenged with SZ-3 exhibit larger mechanocurrents than under control conditions (Fig. 3G,H).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

SZ-3 increases cell stiffness and mechanosensitivity in MCC13 cells. A, Chemical representation of the cofilin inhibitor SZ-3. B, Diagram describing the effect of cofilin inhibition by SZ-3 on actin filaments. Made with BioRender.com. C, Western blot of soluble and insoluble actin (G and F, respectively) of control (DMSO) or SZ-3 (10 µM)-treated MCC13 cells. D, G-/F-actin ratios. Error bars indicate mean ± SD. Two-tailed unpaired t test (t = 2.34). E, Young’s modulus data points are presented as a swarmplot for control (DMSO) or SZ-3 (10 µM)-treated MCC13 cells. Two-tailed Mann–Whitney test (U = 2,509.5). F, Young’s modulus data points are presented as a swarmplot for control (EtOH) and latrunculin A-treated MCC13 cells. Two-tailed Mann–Whitney test (U = 4,596.5). G, Representative whole-cell patch–clamp recordings elicited by mechanical stimulation (−60 mV) of control (DMSO) or SZ-3 (10 µM)-treated MCC13 cells. H, Current densities elicited by maximum displacement of control (DMSO) or SZ-3 (10 µM)-treated MCC13 cells. Error bars indicate mean ± SD. Two-tailed Mann–Whitney test (U = 7). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

To extend our findings to a human neuronal model, we utilized hiPSC-derived sensory neurons generated using the Anatomic Incorporated differentiation kit. In an earlier study, we demonstrated that these in vitro-derived sensory neurons display mechanocurrents characteristic of PIEZO2 (Romero et al., 2023). In these neurons, we also found that SZ-3 enhances PIEZO2 currents while decreasing the displacement threshold (Fig. 4A–C). We have previously shown that DRG neurons from Ube3am–/p+ mice exhibit reduced mechanoexcitability (Romero et al., 2023). Hence, we tested whether SZ-3 could enhance mechanoexcitability in hiPSC-derived sensory neurons. Indeed, neurons treated with SZ-3 required smaller indentation steps (≤6 µm) to elicit mechanically activated action potentials compared with untreated neurons (Fig. 4D,E). Our results demonstrate that SZ-3 increases cell stiffness, mechanocurrents, and PIEZO2 function as well as enhances mechanical excitability in hiPSC-derived sensory neurons.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Cofilin inhibition increases PIEZO2 function and mechanoexcitability in human-derived sensory neurons. A, Representative whole-cell patch–clamp recordings of currents elicited by mechanical stimulation (−60 mV) of control (DMSO) or SZ-3 (5 µM)-treated iPSC–derived sensory neurons. B, Current densities elicited by maximum displacement of control (DMSO) or SZ-3 (5 µM)-treated iPSC–derived neurons. Error bars indicate mean ± SD. Two-tailed unpaired t test (t = 2.92). C, Boxplots show the displacement thresholds required to elicit mechanocurrents in control (DMSO) or SZ-3 (5 µM)-treated iPSC–derived neurons. Two-tailed unpaired t test (t = 2.2). D, Current-clamp recordings of membrane potential changes elicited by indentation of control (DMSO) or SZ-3 (5 µM)-treated iPSC-derived neurons. E, Membrane potential peak versus mechanical indentation of independent iPSC-derived neurons. At the top, boxplots show the displacement threshold required to elicit an action potential in these neurons. Two-tailed Mann–Whitney test (U = 0). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

SZ-3 recovers Ube3am–/p+ neuronal mechanocurrents and mechanoexcitability

We previously showed that, in a mouse model of AS (Ube3am–/p+), DRG neurons exhibit reduced mechanocurrents and mechanoexcitability (Romero et al., 2023). Cultured Ube3am–/p+ mouse DRG neurons challenged with SZ-3 displayed increased mechanocurrents (including the rapidly inactivating currents assigned to PIEZO2) that are reminiscent of those from wild-type (WT) littermate neurons (Fig. 5A,B). The SZ-3 treatment did not affect the distribution of DRG neurons exhibiting rapidly inactivating, intermediate, and slow mechanocurrents (Fig. 5C). Moreover, the displacement threshold required to elicit mechanocurrents in the Ube3am–/p+ neurons was lower after SZ-3 treatment compared with the control and similar to the DRG neurons of WT mice (Fig. 5D). Similarly, DRG neurons from Ube3am–/p+ mice, in the presence of SZ-3, required smaller indentation steps (≤10µm) to trigger action potentials, akin to WT, whereas untreated neurons required larger indentations (≥12 µm; Fig. 5E,F). Overall, we demonstrate that stabilizing F-actin with SZ-3 restores mechanocurrents and mechanoexcitability in DRG neurons of an AS mouse model.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

SZ-3 treatment increases Ube3am–/p+ mouse mechanocurrents and mechanoexcitability. A, Representative whole-cell patch–clamp recording elicited by mechanical stimulation (−60 mV) of rapidly, intermediate, and slowly inactivating currents of WT, control (DMSO) or SZ-3 (5 µM)-treated Ube3am–/p+ DRG neurons. B, Current densities elicited by maximum displacement of DRG neurons, classified by their time constant of inactivation. Error bars indicate mean ± SD. Kruskal–Wallis (H = 38.71; p = 5.56−6) and Dunn’s multiple-comparison test. C, Proportions of WT, control (DMSO), or SZ-3 (5 µM for 18 h)-treated Ube3am–/p+ DRG neurons classified by their time constant of inactivation of mechano-activated currents elicited by maximum displacement. χ2 test (χ2 = 0.22; p = 0.99). D, Displacement thresholds required to elicit mechanocurrents in WT, control (DMSO), or SZ-3(5 µM)-treated Ube3am–/p+ DRG neurons. Error bars indicate mean ± SD. Kruskal–Wallis (H = 9.25; p = 0.0098) and Dunn’s multiple-comparison test. E, Representative current-clamp recordings of membrane potential changes elicited by mechanical stimulation of WT, control (DMSO), or SZ-3 (5 µM)-treated Ube3am–/p+ DRG neurons. F, Membrane potential peak versus mechanical indentation of independent WT, control (DMSO), or SZ-3 (5 µM)-treated Ube3am–/p+ DRG neurons. At the top, boxplots show the displacement threshold required to elicit an action potential in these neurons. Boxplots show mean (square), median (bisecting line), bounds of box (75th to 25th percentiles), outlier range with 1.5 coefficient (whiskers), and minimum and maximum data points. One-way ANOVA (F = 14.52; p = 3.87−5) with Tukey’s multiple-comparison test. n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

SZ-3 increases glutamate-dependent currents in Ube3am–/p+ hippocampal neuron cultures

AMPA receptors are expressed in hippocampal neurons, where they are essential for glutamate-dependent excitatory neurotransmission and spatial learning (Conboy and Sandi, 2010; Keifer and Zheng, 2010; Kamalova and Nakagawa, 2021; Yao et al., 2022). Previous work demonstrated that AMPA receptor currents are decreased in hippocampal neurons from a mouse model of AS (Greer et al., 2010). Importantly, Gu et al. showed that cofilin activity and actin dynamics influence AMPA receptor trafficking and function in rat hippocampal neurons (Gu et al., 2010). Therefore, we hypothesized that inhibiting cofilin activity may enhance AMPA function in the CNS, akin to PIEZO2 in the peripheral nervous system. To assess AMPA receptor function, we recorded AMPA currents in primary hippocampal neurons from Ube3am–/p+ mice and their WT littermates treated with SZ-3 (Fig. 6A,B) while inhibiting NMDA receptors (using Mg2+ and 2-amino-5-phosphonovaleric acid). We found that SZ-3 significantly enhanced glutamate-dependent AMPA currents (Fig. 6A,B). Control experiments in HEK293 cells transiently expressing NMDA receptors, TRPV1, TRPA1, and ASIC1a, demonstrated that SZ-3 did not alter their currents under similar experimental conditions (Fig. 6C,D; Fig. 7). These results suggest that SZ-3 does not have a broad effect on ion channel function. Together, our findings suggest that inhibiting cofilin activity may enhance AMPA receptor function in the context of AS.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

SZ-3 increases glutamate-activated currents in hippocampal neurons from Ube3am–/p+ mice. A, Representative traces of whole-cell patch–clamp recordings, elicited by application of 10 mM glutamate in control (vehicle) or SZ-3 (5 µM; 3 h)-treated primary hippocampal neurons from WT or Ube3am–/p+ mice. B, Current densities of control (vehicle) or SZ-3 (5 µM; 3 h)-treated primary hippocampal neurons from WT or Ube3am–/p+ mice, elicited by 10 mM glutamate. Kruskal–Wallis (H = 32.4; p = 4.33−8) and Dunn’s multiple-comparison test. Error bars indicate mean ± SD. C, Representative whole-cell patch–clamp recordings, elicited by glutamate and glycine (1 mM), of NMDA receptors transfected in HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). D, Current densities, elicited by glutamate and glycine (1 mM), of NMDA receptors transfected in HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). Error bars indicate mean ± SD. Two-tailed Mann–Whitney test (U = 368; p = 0.726). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

SZ-3 does not modulate TRPV1, TRPA1, or ASIC1a activity. A, Representative whole-cell patch–clamp recordings elicited by capsaicin (500 nM and 10 µM) of rTRPV1-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). B, Normalized capsaicin response (500 nM/10 µM) of rTRPV1-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). Error bars indicate mean ± SD. Two-tailed unpaired t test (t = 0.464). C, Representative whole-cell patch–clamp recordings elicited by AITC (5 µM and 100 µM) of hTRPA1-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). D, Normalized AITC response (5 µM/100 µM) of hTRPA1-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). Error bars indicate mean ± SD. Two-tailed unpaired t test (t = −0.033). E, Representative whole-cell patch–clamp recordings, elicited by acidic pH, of rASIC1a-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). F, Current densities, elicited by acidic pH, of rASIC1a-transfected HEK293 cells treated with control (DMSO) or SZ-3 (5 µM). Error bars indicate mean ± SD. Two-way ANOVA with Tukey’s multiple-comparison test (F = 0.078; p = 0.781). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

Cofilin inhibition improves Ube3am–/p+ mice rotarod and T-maze performances

Individuals with AS and the corresponding mouse models exhibit impaired motor coordination and learning disabilities (Tan et al., 2011; Larson et al., 2015; Buiting et al., 2016; Wheeler et al., 2017). The accelerating rotarod is a widely used and reliable assay for evaluating rodent motor coordination (Jiang et al., 2010; Born et al., 2017; Sonzogni et al., 2018; Rotaru et al., 2020; Fig. 8A, bottom panel). While we previously found that a linoleic acid-enriched diet ameliorated gait ataxia in a mouse model of AS, it did not enhance rotarod performance. Since the recovery of peripheral mechanosensory function was insufficient to improve rotarod performance in AS mice (Romero et al., 2023), we hypothesized that inhibiting cofilin in the CNS might improve it. Hence, we administered daily intraperitoneal injections of SZ-3 or the vehicle for 10 consecutive days (Fig. 8A, top panel). Remarkably, SZ-3-treated Ube3am—/p+ mice exhibited improved rotarod performance compared with those receiving the vehicle treatment or WT mice treated with SZ-3 (Fig. 8B,C).

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

SZ-3 improves Ube3am–/p+ mouse rotarod and T-maze performance. A, Diagram depicting the experimental timeline and rotarod behavioral assay. Made with BioRender.com. B, Latency to fall changes before and after 10 d of vehicle or SZ-3 treatment of WT mice. Data samples are paired. Paired Wilcoxon signed-rank test for the vehicle group (W = 6). Two-tailed paired t test for the AZ-3 group (t = 0.392). C, Latency to fall changes before and after 10 d of vehicle or SZ-3 treatment of Ube3am–/p+ mice. Data samples are paired. Two-tailed paired t test (t = 0.24 for vehicle and t = 5.22 for SZ-3). D, Diagram depicting the experimental timeline and T-maze behavioral assay. Made with BioRender.com. E, Percentage alternation at Day 7 of the vehicle or SZ-3 treatment of Ube3am–/p+ mice. Error bars indicate mean ± SD. Two-tailed Mann–Whitney test (U = 6). E, Percentage alternation at Day 12 of the vehicle or SZ-3 treatment of Ube3am–/p+ mice. Error bars indicate mean ± SD. Two-tailed Mann–Whitney test (U = 3.5). F, Diagram depicting the experimental timeline and the von Frey behavioral assay. Made with BioRender.com. G, Withdrawal thresholds elicited with von Frey filaments before and after 10 d of vehicle or SZ-3 treatment of WT mice. Paired Wilcoxon signed-rank test for the vehicle group (W = 12.5). Two-tailed paired t test for the AZ-3 group (t = 0.326). n is denoted in each panel. Post hoc p values are denoted in the corresponding panels.

T-mazes are standard behavioral assays used to assess the cognitive abilities (e.g., spatial learning and memory) of rodents (Deacon and Rawlins, 2006; Fig. 8D, bottom panel). We have previously utilized this behavioral test and demonstrated that SZ-3 improves cognitive function in a mouse model (WT) of intracerebral hemorrhage (Almarghalani et al., 2024). Since T-maze alternation is sensitive to hippocampal dysfunction, we tested whether SZ-3 treatment could improve the performance of this task. Notably, after 7 consecutive days of SZ-3 injection, Ube3am–/p+ mice exhibited a significant increase in T-maze alternation (Fig. 8D,E). This improvement in spatial learning and memory persisted when the test was repeated 2 d after the 10 d treatment was completed (Fig. 8E). Control experiments in WT mice showed that SZ-3 did not affect touch sensitivity, as determined by von Frey assays (Fig. 8F,G). Overall, our results highlight the role of actin-binding proteins in motor and learning disabilities in the context of AS.

Discussion

Our study sheds light on how cofilin modulates ion channel function, underscoring the therapeutic potential of targeting actin-binding proteins in AS. Here, we demonstrate that acute cofilin activation using optogenetics decreases mechanocurrents, while genetic and pharmacological (with the first-in-class cofilin inhibitor SZ-3) inhibition restores PIEZO2 currents and mechanoexcitability in AS. Additionally, we show that SZ-3 restores AMPA receptor function in cultured hippocampal neurons and enhances motor coordination and cognitive function in a mouse model of AS.

The cytoskeleton continuously regulates the function of membrane proteins, including ion channels (Janmey et al., 1998). Proper actin turnover and organization are critical for maintaining channel mobility and activity (Spence and Soderling, 2015). Thus, proteins that govern the dynamic assembly and disassembly of the actin cytoskeleton (e.g., cofilin) may influence channel function under basal conditions, including those involving PIEZO2 and AMPA receptors. Previously, we demonstrated that loss of UBE3A expression leads to elevated cofilin levels, reduced F-actin, and impaired PIEZO2 mechanocurrents (Romero et al., 2023). Several studies have shown that force-from-filament (FFF) mechanisms modulate the function of PIEZO channels, and disruption of cytoskeletal components—such as actin, tubulin, or β-catenin—leads to impaired mechanotransduction (Eijkelkamp et al., 2013; Romero et al., 2020; Verkest et al., 2022). This functional interplay may offer a compensatory advantage: stabilizing the cytoskeleton could help mitigate the consequences of pathogenic PIEZO2 LOF phenotypes. Indeed, one of our key findings is that the cofilin inhibitor SZ-3 restores mechanocurrents (including those of PIEZO2) and mechanoexcitability in DRG neurons from an AS mouse model by increasing their F-actin and Young’s modulus. Notably, by optogenetically severing F-actin with Z-lock cofilin, we could identify the role of intact filaments in mechanotransduction. This disruption occurred within minutes, faster than membrane protein trafficking, which generally occurs over larger timescales. Our data support the notion that actin dynamics acutely fine-tune the function of mechanosensitive ion channels. This, however, does not contradict the force-from-lipids (FFL) principle but instead suggests that FFF and FFL mechanisms act together to modulate mechanosensitivity, as previously proposed (Goodman et al., 2023).

Our earlier work showed that prolonged disruption of F-actin with latrunculin A—mimicking the effects of continuously elevated cofilin levels in the DRG neurons of Ube3a-deficient mice—leads to reduced membrane expression of PIEZO2 (Romero et al., 2023). Therefore, our results suggest that in AS, actin filament disorganization may impair both the long-term trafficking of PIEZO2 to the plasma membrane and the rapid, second-scale neuronal mechanics required for precise mechanosensation. A previous work showed that hippocampal neurons have a reduction in F-actin in AS (Baudry et al., 2012). Actin, the primary cytoskeletal element in mature synapses, is essential for synaptogenesis, maintenance, and plasticity (Shaw and Koleske, 2021). AMPA receptor density is reduced in hippocampal excitatory synapses, a defect that may contribute to the cognitive impairment observed in AS (Greer et al., 2010). Our findings confirm impaired AMPA receptor function in AS and identify cofilin inhibition as a strategy to improve channel function.

Motor and cognitive deficits are among the most common and debilitating aspects of AS (Margolis et al., 2015). Our finding that SZ-3 ameliorates these phenotypes in an AS mouse model highlights potential alternative therapeutic targets beyond UBE3A. We have recently shown that SZ-3 possesses favorable pharmacological and pharmacokinetic profiles for treating neurodegenerative disorders (Alsegiani and Shah, 2023). Given the widespread expression of cofilin across nearly all cell types, systemic administration may be suboptimal. Future studies should focus on developing targeted delivery strategies to improve the actin cytoskeleton solely in mechanoreceptor and hippocampal neurons. Although SZ-3 enhanced PIEZO2 channel and AMPA receptor function, as well as improved performance in the rotarod and T-maze tests, our study does not exclude the possibility that its behavioral effects may also involve mechanisms beyond the enhancement of neuronal ion channel function. Nevertheless, our work provides proof of concept that inhibiting cofilin is a potential strategy for improving motor coordination and spatial learning in AS.

Footnotes

  • We thank Drs. Briar Bell, Leidys French-Pacheco, and Angela Jaramillo Granada for critically reading the manuscript and for technical support. We utilized the Atomic Force Microscopy Core Facility at UTHealth Houston and thank Dr. Ana María Saske for her technical support. We thank Drs. Rebeca Caires and Rakesh Kumar for their early contributions to the mouse work. This work was supported by the National Institutes of Health R35GM122528 to V.J., R01NS112642 to Z.A.S., R35GM153208 to V.V., R35GM149218 to J.F.C.-M, and R01NS131184 to J.F.C.-M and V.V.

  • The authors declare no competing financial interests.

  • Correspondence should be addressed to Valeria Vásquez at valeria.vasquez{at}uth.tmc.edu or Julio F. Cordero-Morales at julio.cordero{at}uth.tmc.edu.

This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license, which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

References

  1. ↵
    1. Aiken J,
    2. Holzbaur ELF
    (2021) Cytoskeletal regulation guides neuronal trafficking to effectively supply the synapse. Curr Biol 31:R633–R650. https://doi.org/10.1016/j.cub.2021.02.024
    OpenUrlCrossRefPubMed
  2. ↵
    1. Alaqel SI,
    2. Dlamini S,
    3. Almarghalani DA,
    4. Shettigar A,
    5. Alhadidi Q,
    6. Kodithuwakku SH,
    7. Stary C,
    8. Tillekeratne LMV,
    9. Shah ZA
    (2022) Synthesis and development of a novel first-in-class cofilin inhibitor for neuroinflammation in hemorrhagic brain injury. ACS Chem Neurosci 13:1014–1029. https://doi.org/10.1021/acschemneuro.2c00010
    OpenUrlCrossRefPubMed
  3. ↵
    1. Almarghalani DA,
    2. Bahader GA,
    3. Ali M,
    4. Tillekeratne LMV,
    5. Shah ZA
    (2024) Cofilin inhibitor improves neurological and cognitive functions after intracerebral hemorrhage by suppressing endoplasmic reticulum stress related-neuroinflammation. Pharmaceuticals (Basel) 17:1–19. https://doi.org/10.3390/ph17010114
    OpenUrl
  4. ↵
    1. Alsegiani AS,
    2. Shah ZA
    (2023) Development and validation of LC-MS/MS method for determining the metabolic stability, pharmacokinetics, and dose proportionality of a novel anti-inflammatory cofilin inhibitor. J Pharm Biomed Anal 226:115258. https://doi.org/10.1016/j.jpba.2023.115258
    OpenUrlPubMed
  5. ↵
    1. Bahader GA,
    2. James AW,
    3. Almarghalani DA,
    4. Shah ZA
    (2023) Cofilin inhibitor protects against traumatic brain injury-induced oxidative stress and neuroinflammation. Biology (Basel) 12:1–16. https://doi.org/10.3390/biology12040630
    OpenUrl
  6. ↵
    1. Bamburg JR
    (1999) Proteins of the ADF/cofilin family: essential regulators of actin dynamics. Annu Rev Cell Dev Biol 15:185–230. https://doi.org/10.1146/annurev.cellbio.15.1.185
    OpenUrlCrossRefPubMed
  7. ↵
    1. Baudry M,
    2. Kramar E,
    3. Xu X,
    4. Zadran H,
    5. Moreno S,
    6. Lynch G,
    7. Gall C,
    8. Bi X
    (2012) Ampakines promote spine actin polymerization, long-term potentiation, and learning in a mouse model of Angelman syndrome. Neurobiol Dis 47:210–215. https://doi.org/10.1016/j.nbd.2012.04.002
    OpenUrlCrossRefPubMed
  8. ↵
    1. Born HA,
    2. Dao AT,
    3. Levine AT,
    4. Lee WL,
    5. Mehta NM,
    6. Mehra S,
    7. Weeber EJ,
    8. Anderson AE
    (2017) Strain-dependence of the Angelman syndrome phenotypes in Ube3a maternal deficiency mice. Sci Rep 7:8451. https://doi.org/10.1038/s41598-017-08825-x
    OpenUrlCrossRefPubMed
  9. ↵
    1. Buiting K,
    2. Williams C,
    3. Horsthemke B
    (2016) Angelman syndrome - insights into a rare neurogenetic disorder. Nat Rev Neurol 12:584–593. https://doi.org/10.1038/nrneurol.2016.133
    OpenUrlCrossRefPubMed
  10. ↵
    1. Carrillo E,
    2. Bhatia NK,
    3. Akimzhanov AM,
    4. Jayaraman V
    (2020) Activity dependent inhibition of AMPA receptors by Zn(2). J Neurosci 40:8629–8636. https://doi.org/10.1523/JNEUROSCI.1481-20.2020
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Carrillo E,
    2. Gonzalez CU,
    3. Berka V,
    4. Jayaraman V
    (2021) Delta glutamate receptors are functional glycine- and d-serine-gated cation channels in situ. Sci Adv 7:eabk2200. https://doi.org/10.1126/sciadv.abk2200
    OpenUrlCrossRefPubMed
  12. ↵
    1. Chang EH,
    2. Savage MJ,
    3. Flood DG,
    4. Thomas JM,
    5. Levy RB,
    6. Mahadomrongkul V,
    7. Shirao T,
    8. Aoki C,
    9. Huerta PT
    (2006) AMPA receptor downscaling at the onset of Alzheimer’s disease pathology in double knockin mice. Proc Natl Acad Sci U S A 103:3410–3415. https://doi.org/10.1073/pnas.0507313103
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Chesler AT, et al.
    (2016) The role of PIEZO2 in human mechanosensation. N Engl J Med 375:1355–1364. https://doi.org/10.1056/NEJMoa1602812
    OpenUrlCrossRefPubMed
  14. ↵
    1. Cingolani LA,
    2. Goda Y
    (2008) Actin in action: the interplay between the actin cytoskeleton and synaptic efficacy. Nat Rev Neurosci 9:344–356. https://doi.org/10.1038/nrn2373
    OpenUrlCrossRefPubMed
  15. ↵
    1. Clayton-Smith J,
    2. Laan L
    (2003) Angelman syndrome: a review of the clinical and genetic aspects. J Med Genet 40:87–95. https://doi.org/10.1136/jmg.40.2.87
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Conboy L,
    2. Sandi C
    (2010) Stress at learning facilitates memory formation by regulating AMPA receptor trafficking through a glucocorticoid action. Neuropsychopharmacology 35:674–685. https://doi.org/10.1038/npp.2009.172
    OpenUrlCrossRefPubMed
  17. ↵
    1. Coste B,
    2. Mathur J,
    3. Schmidt M,
    4. Earley TJ,
    5. Ranade S,
    6. Petrus MJ,
    7. Dubin AE,
    8. Patapoutian A
    (2010) Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science 330:55–60. https://doi.org/10.1126/science.1193270
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Deacon RM,
    2. Rawlins JN
    (2006) T-maze alternation in the rodent. Nat Protoc 1:7–12. https://doi.org/10.1038/nprot.2006.2
    OpenUrlCrossRefPubMed
  19. ↵
    1. Diering GH,
    2. Huganir RL
    (2018) The AMPA receptor code of synaptic plasticity. Neuron 100:314–329. https://doi.org/10.1016/j.neuron.2018.10.018
    OpenUrlCrossRefPubMed
  20. ↵
    1. Ding R, et al.
    (2016) Changes in hippocampal AMPA receptors and cognitive impairments in chronic ketamine addiction models: another understanding of ketamine CNS toxicity. Sci Rep 6:38771. https://doi.org/10.1038/srep38771
    OpenUrlCrossRefPubMed
  21. ↵
    1. Duis J, et al.
    (2022) A multidisciplinary approach and consensus statement to establish standards of care for Angelman syndrome. Mol Genet Genomic Med 10:e1843. https://doi.org/10.1002/mgg3.1843
    OpenUrl
  22. ↵
    1. Eijkelkamp N, et al.
    (2013) A role for Piezo2 in EPAC1-dependent mechanical allodynia. Nat Commun 4:1682. https://doi.org/10.1038/ncomms2673
    OpenUrlCrossRefPubMed
  23. ↵
    1. Fink JJ,
    2. Robinson TM,
    3. Germain ND,
    4. Sirois CL,
    5. Bolduc KA,
    6. Ward AJ,
    7. Rigo F,
    8. Chamberlain SJ,
    9. Levine ES
    (2017) Disrupted neuronal maturation in Angelman syndrome-derived induced pluripotent stem cells. Nat Commun 8:15038. https://doi.org/10.1038/ncomms15038
    OpenUrlCrossRefPubMed
  24. ↵
    1. Goodman MB,
    2. Haswell ES,
    3. Vásquez V
    (2023) Mechanosensitive membrane proteins: usual and unusual suspects in mediating mechanotransduction. J Gen Physiol 155:1–11. https://doi.org/10.1085/jgp.202213248
    OpenUrl
  25. ↵
    1. Greer PL, et al.
    (2010) The Angelman syndrome protein Ube3A regulates synapse development by ubiquitinating arc. Cell 140:704–716. https://doi.org/10.1016/j.cell.2010.01.026
    OpenUrlCrossRefPubMed
  26. ↵
    1. Gu J, et al.
    (2010) ADF/cofilin-mediated actin dynamics regulate AMPA receptor trafficking during synaptic plasticity. Nat Neurosci 13:1208–1215. https://doi.org/10.1038/nn.2634
    OpenUrlCrossRefPubMed
  27. ↵
    1. Henley JM,
    2. Wilkinson KA
    (2013) AMPA receptor trafficking and the mechanisms underlying synaptic plasticity and cognitive aging. Dialogues Clin Neurosci 15:11–27. https://doi.org/10.31887/DCNS.2013.15.1/jhenley
    OpenUrlCrossRefPubMed
  28. ↵
    1. Henley JM,
    2. Wilkinson KA
    (2016) Synaptic AMPA receptor composition in development, plasticity and disease. Nat Rev Neurosci 17:337–350. https://doi.org/10.1038/nrn.2016.37
    OpenUrlCrossRefPubMed
  29. ↵
    1. Ho J,
    2. Tumkaya T,
    3. Aryal S,
    4. Choi H,
    5. Claridge-Chang A
    (2019) Moving beyond p values: data analysis with estimation graphics. Nat Methods 16:565–566. https://doi.org/10.1038/s41592-019-0470-3
    OpenUrlCrossRefPubMed
  30. ↵
    1. Huang TY,
    2. DerMardirossian C,
    3. Bokoch GM
    (2006) Cofilin phosphatases and regulation of actin dynamics. Curr Opin Cell Biol 18:26–31. https://doi.org/10.1016/j.ceb.2005.11.005
    OpenUrlCrossRefPubMed
  31. ↵
    1. Janmey PA,
    2. Kas J,
    3. Shah JV,
    4. Allen PG,
    5. Tang JX
    (1998) Cytoskeletal networks and filament bundles: regulation by proteins and polycations. Biol Bull 194:334–335; discussion 335–336. https://doi.org/10.2307/1543105
    OpenUrlCrossRefPubMed
  32. ↵
    1. Jiang YH,
    2. Armstrong D,
    3. Albrecht U,
    4. Atkins CM,
    5. Noebels JL,
    6. Eichele G,
    7. Sweatt JD,
    8. Beaudet AL
    (1998) Mutation of the Angelman ubiquitin ligase in mice causes increased cytoplasmic p53 and deficits of contextual learning and long-term potentiation. Neuron 21:799–811. https://doi.org/10.1016/S0896-6273(00)80596-6
    OpenUrlCrossRefPubMed
  33. ↵
    1. Jiang YH,
    2. Pan Y,
    3. Zhu L,
    4. Landa L,
    5. Yoo J,
    6. Spencer C,
    7. Lorenzo I,
    8. Brilliant M,
    9. Noebels J,
    10. Beaudet AL
    (2010) Altered ultrasonic vocalization and impaired learning and memory in Angelman syndrome mouse model with a large maternal deletion from Ube3a to Gabrb3. PLoS One 5:e12278. https://doi.org/10.1371/journal.pone.0012278
    OpenUrlCrossRefPubMed
  34. ↵
    1. Judson MC, et al.
    (2016) GABAergic neuron-specific loss of Ube3a causes angelman syndrome-like EEG abnormalities and enhances seizure susceptibility. Neuron 90:56–69. https://doi.org/10.1016/j.neuron.2016.02.040
    OpenUrlCrossRefPubMed
  35. ↵
    1. Kamalova A,
    2. Nakagawa T
    (2021) AMPA receptor structure and auxiliary subunits. J Physiol 599:453–469. https://doi.org/10.1113/JP278701
    OpenUrlCrossRefPubMed
  36. ↵
    1. Keifer J,
    2. Zheng Z
    (2010) AMPA receptor trafficking and learning. Eur J Neurosci 32:269–277. https://doi.org/10.1111/j.1460-9568.2010.07339.x
    OpenUrlCrossRefPubMed
  37. ↵
    1. Kishino T,
    2. Lalande M,
    3. Wagstaff J
    (1997) UBE3A/E6-AP mutations cause Angelman syndrome. Nat Genet 15:70–73. https://doi.org/10.1038/ng0197-70
    OpenUrlCrossRefPubMed
  38. ↵
    1. Knoll JH,
    2. Nicholls RD,
    3. Magenis RE,
    4. Graham JM Jr.,
    5. Lalande M,
    6. Latt SA
    (1989) Angelman and Prader-Willi syndromes share a common chromosome 15 deletion but differ in parental origin of the deletion. Am J Med Genet 32:285–290. https://doi.org/10.1002/ajmg.1320320235
    OpenUrlCrossRefPubMed
  39. ↵
    1. Larson AM,
    2. Shinnick JE,
    3. Shaaya EA,
    4. Thiele EA,
    5. Thibert RL
    (2015) Angelman syndrome in adulthood. Am J Med Genet A 167A:331–344. https://doi.org/10.1002/ajmg.a.36864
    OpenUrlCrossRefPubMed
  40. ↵
    1. Lopez SJ,
    2. Dunaway K,
    3. Islam MS,
    4. Mordaunt C,
    5. Vogel Ciernia A,
    6. Meguro-Horike M,
    7. Horike SI,
    8. Segal DJ,
    9. LaSalle JM
    (2017) UBE3A-mediated regulation of imprinted genes and epigenome-wide marks in human neurons. Epigenetics 12:982–990. https://doi.org/10.1080/15592294.2017.1376151
    OpenUrlCrossRefPubMed
  41. ↵
    1. Lopez SJ,
    2. Segal DJ,
    3. LaSalle JM
    (2018) UBE3A: an E3 ubiquitin ligase with genome-wide impact in neurodevelopmental disease. Front Mol Neurosci 11:476. https://doi.org/10.3389/fnmol.2018.00476
    OpenUrlPubMed
  42. ↵
    1. Maciver SK,
    2. Hussey PJ
    (2002) The ADF/cofilin family: actin-remodeling proteins. Genome Biol 3:reviews3007. https://doi.org/10.1186/gb-2002-3-5-reviews3007
    OpenUrlCrossRefPubMed
  43. ↵
    1. Margolis SS,
    2. Sell GL,
    3. Zbinden MA,
    4. Bird LM
    (2015) Angelman syndrome. Neurotherapeutics 12:641–650. https://doi.org/10.1007/s13311-015-0361-y
    OpenUrlCrossRefPubMed
  44. ↵
    1. McCoy ES,
    2. Taylor-Blake B,
    3. Aita M,
    4. Simon JM,
    5. Philpot BD,
    6. Zylka MJ
    (2017) Enhanced nociception in Angelman syndrome model mice. J Neurosci 37:10230–10239. https://doi.org/10.1523/JNEUROSCI.1018-17.2017
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Miura K,
    2. Kishino T,
    3. Li E,
    4. Webber H,
    5. Dikkes P,
    6. Holmes GL,
    7. Wagstaff J
    (2002) Neurobehavioral and electroencephalographic abnormalities in Ube3a maternal-deficient mice. Neurobiol Dis 9:149–159. https://doi.org/10.1006/nbdi.2001.0463
    OpenUrlCrossRefPubMed
  46. ↵
    1. Nagel M,
    2. Chesler AT
    (2022) PIEZO2 ion channels in proprioception. Curr Opin Neurobiol 75:102572. https://doi.org/10.1016/j.conb.2022.102572
    OpenUrlCrossRefPubMed
  47. ↵
    1. Ranade SS, et al.
    (2014) Piezo2 is the major transducer of mechanical forces for touch sensation in mice. Nature 516:121–125. https://doi.org/10.1038/nature13980
    OpenUrlCrossRefPubMed
  48. ↵
    1. Romero LO,
    2. Caires R,
    3. Nickolls AR,
    4. Chesler AT,
    5. Cordero-Morales JF,
    6. Vasquez V
    (2020) A dietary fatty acid counteracts neuronal mechanical sensitization. Nat Commun 11:2997. https://doi.org/10.1038/s41467-020-16816-2
    OpenUrlCrossRefPubMed
  49. ↵
    1. Romero LO, et al.
    (2023) Linoleic acid improves PIEZO2 dysfunction in a mouse model of Angelman syndrome. Nat Commun 14:1167. https://doi.org/10.1038/s41467-023-36818-0
    OpenUrlCrossRefPubMed
  50. ↵
    1. Rotaru DC,
    2. Mientjes EJ,
    3. Elgersma Y
    (2020) Angelman syndrome: from mouse models to therapy. Neuroscience 445:172–189. https://doi.org/10.1016/j.neuroscience.2020.02.017
    OpenUrlCrossRefPubMed
  51. ↵
    1. Sasaki S,
    2. Yui N,
    3. Noda Y
    (2014) Actin directly interacts with different membrane channel proteins and influences channel activities: AQP2 as a model. Biochim Biophys Acta 1838:514–520. https://doi.org/10.1016/j.bbamem.2013.06.004
    OpenUrlCrossRefPubMed
  52. ↵
    1. Scheffner M,
    2. Huibregtse JM,
    3. Vierstra RD,
    4. Howley PM
    (1993) The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 75:495–505. https://doi.org/10.1016/0092-8674(93)90384-3
    OpenUrlCrossRefPubMed
  53. ↵
    1. Shaw JE,
    2. Koleske AJ
    (2021) Functional interactions of ion channels with the actin cytoskeleton: does coupling to dynamic actin regulate NMDA receptors? J Physiol 599:431–441. https://doi.org/10.1113/JP278702
    OpenUrlPubMed
  54. ↵
    1. Sonzogni M,
    2. Wallaard I,
    3. Santos SS,
    4. Kingma J,
    5. du Mee D,
    6. van Woerden GM,
    7. Elgersma Y
    (2018) A behavioral test battery for mouse models of Angelman syndrome: a powerful tool for testing drugs and novel Ube3a mutants. Mol Autism 9:47. https://doi.org/10.1186/s13229-018-0231-7
    OpenUrlCrossRefPubMed
  55. ↵
    1. Spence EF,
    2. Soderling SH
    (2015) Actin out: regulation of the synaptic cytoskeleton. J Biol Chem 290:28613–28622. https://doi.org/10.1074/jbc.R115.655118
    OpenUrlAbstract/FREE Full Text
  56. ↵
    1. Stone OJ, et al.
    (2019) Optogenetic control of cofilin and alphaTAT in living cells using Z-lock. Nat Chem Biol 15:1183–1190. https://doi.org/10.1038/s41589-019-0405-4
    OpenUrlCrossRefPubMed
  57. ↵
    1. Szczot M,
    2. Nickolls AR,
    3. Lam RM,
    4. Chesler AT
    (2021) The form and function of PIEZO2. Annu Rev Biochem 90:507–534. https://doi.org/10.1146/annurev-biochem-081720-023244
    OpenUrlCrossRefPubMed
  58. ↵
    1. Tan WH, et al.
    (2011) Angelman syndrome: mutations influence features in early childhood. Am J Med Genet A 155A:81–90. https://doi.org/10.1002/ajmg.a.33775
    OpenUrlCrossRef
  59. ↵
    1. Tucci V,
    2. Isles AR,
    3. Kelsey G,
    4. Ferguson-Smith AC,
    5. Erice Imprinting G
    (2019) Genomic imprinting and physiological processes in mammals. Cell 176:952–965. https://doi.org/10.1016/j.cell.2019.01.043
    OpenUrlCrossRefPubMed
  60. ↵
    1. Verkest C,
    2. Schaefer I,
    3. Nees TA,
    4. Wang N,
    5. Jegelka JM,
    6. Taberner FJ,
    7. Lechner SG
    (2022) Intrinsically disordered intracellular domains control key features of the mechanically-gated ion channel PIEZO2. Nat Commun 13:1365. https://doi.org/10.1038/s41467-022-28974-6
    OpenUrlCrossRefPubMed
  61. ↵
    1. Walz NC,
    2. Baranek GT
    (2006) Sensory processing patterns in persons with Angelman syndrome. Am J Occup Ther 60:472–479. https://doi.org/10.5014/ajot.60.4.472
    OpenUrlCrossRefPubMed
  62. ↵
    1. Wang J,
    2. Jiang J,
    3. Yang X,
    4. Zhou G,
    5. Wang L,
    6. Xiao B
    (2022) Tethering Piezo channels to the actin cytoskeleton for mechanogating via the cadherin-beta-catenin mechanotransduction complex. Cell Rep 38:110342. https://doi.org/10.1016/j.celrep.2022.110342
    OpenUrlCrossRefPubMed
  63. ↵
    1. Wheeler AC,
    2. Sacco P,
    3. Cabo R
    (2017) Unmet clinical needs and burden in Angelman syndrome: a review of the literature. Orphanet J Rare Dis 12:164. https://doi.org/10.1186/s13023-017-0716-z
    OpenUrlCrossRefPubMed
  64. ↵
    1. Wioland H,
    2. Guichard B,
    3. Senju Y,
    4. Myram S,
    5. Lappalainen P,
    6. Jegou A,
    7. Romet-Lemonne G
    (2017) ADF/cofilin accelerates actin dynamics by severing filaments and promoting their depolymerization at both ends. Curr Biol 27:1956–1967 e1957. https://doi.org/10.1016/j.cub.2017.05.048
    OpenUrlCrossRefPubMed
  65. ↵
    1. Woo SH, et al.
    (2014) Piezo2 is required for Merkel-cell mechanotransduction. Nature 509:622–626. https://doi.org/10.1038/nature13251
    OpenUrlCrossRefPubMed
  66. ↵
    1. Woo SH,
    2. Lukacs V,
    3. de Nooij JC,
    4. Zaytseva D,
    5. Criddle CR,
    6. Francisco A,
    7. Jessell TM,
    8. Wilkinson KA,
    9. Patapoutian A
    (2015) Piezo2 is the principal mechanotransduction channel for proprioception. Nat Neurosci 18:1756–1762. https://doi.org/10.1038/nn.4162
    OpenUrlCrossRefPubMed
  67. ↵
    1. Yao D, et al.
    (2022) Hippocampal AMPA receptors mediate the impairment of spatial learning and memory in prenatally stressed offspring rats. J Psychiatr Res 151:17–24. https://doi.org/10.1016/j.jpsychires.2022.03.032
    OpenUrlPubMed
  68. ↵
    1. Zech T,
    2. Calaminus SD,
    3. Machesky LM
    (2012) Actin on trafficking: could actin guide directed receptor transport? Cell Adh Migr 6:476–481. https://doi.org/10.4161/cam.21373
    OpenUrlCrossRefPubMed
Back to top

In this issue

The Journal of Neuroscience: 45 (45)
Journal of Neuroscience
Vol. 45, Issue 45
5 Nov 2025
  • Table of Contents
  • About the Cover
  • Index by author
  • Masthead (PDF)
Email

Thank you for sharing this Journal of Neuroscience article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Cofilin Inhibition Ameliorates PIEZO2 and AMPA Dysfunction in a Mouse Model of Angelman Syndrome
(Your Name) has forwarded a page to you from Journal of Neuroscience
(Your Name) thought you would be interested in this article in Journal of Neuroscience.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
View Full Page PDF
Citation Tools
Cofilin Inhibition Ameliorates PIEZO2 and AMPA Dysfunction in a Mouse Model of Angelman Syndrome
Luis O. Romero, Manisha Bade, Elisa Carrillo, Sonia Paz-López, Syed A. M. Hasan, William James Antonisamy, Vasanthi Jayaraman, Zahoor A. Shah, Valeria Vásquez, Julio F. Cordero-Morales
Journal of Neuroscience 5 November 2025, 45 (45) e0965252025; DOI: 10.1523/JNEUROSCI.0965-25.2025

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Respond to this article
Request Permissions
Share
Cofilin Inhibition Ameliorates PIEZO2 and AMPA Dysfunction in a Mouse Model of Angelman Syndrome
Luis O. Romero, Manisha Bade, Elisa Carrillo, Sonia Paz-López, Syed A. M. Hasan, William James Antonisamy, Vasanthi Jayaraman, Zahoor A. Shah, Valeria Vásquez, Julio F. Cordero-Morales
Journal of Neuroscience 5 November 2025, 45 (45) e0965252025; DOI: 10.1523/JNEUROSCI.0965-25.2025
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Significance Statement
    • Introduction
    • Material and methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • Peer Review
  • PDF

Keywords

  • AMPA receptor
  • Angelman syndrome
  • cofilin
  • hippocampal neurons
  • PIEZO2
  • sensory neurons

Responses to this article

Respond to this article

Jump to comment:

No eLetters have been published for this article.

Related Articles

Cited By...

More in this TOC Section

Research Articles

  • Co-release of GABA and ACh from Medial Olivocochlear Neurons as a Fine Regulatory Mechanism of Cochlear Efferent Inhibition
  • Collapsing Perisomatic Inhibition Leads to Epileptic Fast-Ripple Oscillations Caused by Pseudosynchronous Firing of CA3 Pyramidal Neurons
  • Behavior-Relevant Periodized Neural Representation of Acoustic But Not Tactile Rhythm in Humans
Show more Research Articles

Cellular/Molecular

  • Co-release of GABA and ACh from Medial Olivocochlear Neurons as a Fine Regulatory Mechanism of Cochlear Efferent Inhibition
  • Synaptic Gpr85 influences cerebellar granule cells electrical properties and light-induced behavior in zebrafish.
Show more Cellular/Molecular
  • Home
  • Alerts
  • Follow SFN on BlueSky
  • Visit Society for Neuroscience on Facebook
  • Follow Society for Neuroscience on Twitter
  • Follow Society for Neuroscience on LinkedIn
  • Visit Society for Neuroscience on Youtube
  • Follow our RSS feeds

Content

  • Early Release
  • Current Issue
  • Issue Archive
  • Collections

Information

  • For Authors
  • For Advertisers
  • For the Media
  • For Subscribers

About

  • About the Journal
  • Editorial Board
  • Privacy Notice
  • Contact
  • Accessibility
(JNeurosci logo)
(SfN logo)

Copyright © 2025 by the Society for Neuroscience.
JNeurosci Online ISSN: 1529-2401

The ideas and opinions expressed in JNeurosci do not necessarily reflect those of SfN or the JNeurosci Editorial Board. Publication of an advertisement or other product mention in JNeurosci should not be construed as an endorsement of the manufacturer’s claims. SfN does not assume any responsibility for any injury and/or damage to persons or property arising from or related to any use of any material contained in JNeurosci.