Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Fernanda Laezza is active.

Publication


Featured researches published by Fernanda Laezza.


The Journal of Physiology | 2005

Fibroblast growth factor 14 is an intracellular modulator of voltage‐gated sodium channels

Jun Yang Lou; Fernanda Laezza; Benjamin R. Gerber; Maolei Xiao; Kathryn A. Yamada; Hali Hartmann; Ann Marie Craig; Jeanne M. Nerbonne; David M. Ornitz

Genetic ablation of the fibroblast growth factor (Fgf) 14 gene in mice or a missense mutation in Fgf14 in humans causes ataxia and cognitive deficits. These phenotypes suggest that the neuronally expressed Fgf14 gene is essential for regulating normal neuronal activity. Here, we demonstrate that FGF14 interacts directly with multiple voltage‐gated Na+ (Nav) channel α subunits heterologously expressed in non‐neuronal cells or natively expressed in a murine neuroblastoma cell line. Functional studies reveal that these interactions result in the potent inhibition of Nav channel currents (INa) and in changes in the voltage dependence of channel activation and inactivation. Deletion of the unique amino terminus of the splice variant of Fgf14, Fgf14‐1b, or expression of the splice variant Fgf14‐1a modifies the modulatory effects on INa, suggesting an important role for the amino terminus domain of FGF14 in the regulation of Nav channels. To investigate the function of FGF14 in neurones, we directly expressed Fgf14 in freshly isolated primary rat hippocampal neurones. In these cells, the addition of FGF14‐1a–GFP or FGF14‐1b–GFP increased INa density and shifted the voltage dependence of channel activation and inactivation. In fully differentiated neurones, FGF14‐1a–GFP or FGF14‐1b–GFP preferentially colocalized with endogenous Nav channels at the axonal initial segment, a critical region for action potential generation. Together, these findings implicate FGF14 as a unique modulator of Nav channel activity in the CNS and provide a possible mechanism to explain the neurological phenotypes observed in mice and humans with mutations in Fgf14.


The Journal of Neuroscience | 2007

The FGF14F145S Mutation Disrupts the Interaction of FGF14 with Voltage-Gated Na+ Channels and Impairs Neuronal Excitability

Fernanda Laezza; Benjamin R. Gerber; Jun Yang Lou; Marie A. Kozel; Hali Hartman; Ann Marie Craig; David M. Ornitz; Jeanne M. Nerbonne

Fibroblast growth factor 14 (FGF14) belongs to the intracellular FGF homologous factor subfamily of FGF proteins (iFGFs) that are not secreted and do not activate tyrosine kinase receptors. The iFGFs, however, have been shown to interact with the pore-forming (α) subunits of voltage-gated Na+ (Nav) channels. The neurological phenotypes seen in Fgf14−/− mice and the identification of an FGF14 missense mutation (FGF14F145S) in a Dutch family presenting with cognitive impairment and spinocerebellar ataxia suggest links between FGF14 and neuronal functioning. Here, we demonstrate that the expression of FGF14F145S reduces Nav α subunit expression at the axon initial segment, attenuates Nav channel currents, and reduces the excitability of hippocampal neurons. In addition, and in contrast with wild-type FGF14, FGF14F145S does not interact directly with Nav channel α subunits. Rather, FGF14F145S associates with wild-type FGF14 and disrupts the interaction between wild-type FGF14 and Nav α subunits, suggesting that the mutant FGF14F145S protein acts as a dominant negative, interfering with the interaction between wild-type FGF14 and Nav channel α subunits and altering neuronal excitability.


Journal of Biological Chemistry | 2009

Crystal Structure of a Fibroblast Growth Factor Homologous Factor (FHF) Defines a Conserved Surface on FHFs for Binding and Modulation of Voltage-gated Sodium Channels

Regina Goetz; Katarzyna Dover; Fernanda Laezza; Nataly Shtraizent; Xiao Huang; Dafna Tchetchik; Anna V. Eliseenkova; Chong Feng Xu; Thomas A. Neubert; David M. Ornitz; Mitchell Goldfarb; Moosa Mohammadi

Voltage-gated sodium channels (Nav) produce sodium currents that underlie the initiation and propagation of action potentials in nerve and muscle cells. Fibroblast growth factor homologous factors (FHFs) bind to the intracellular C-terminal region of the Nav α subunit to modulate fast inactivation of the channel. In this study we solved the crystal structure of a 149-residue-long fragment of human FHF2A which unveils the structural features of the homology core domain of all 10 human FHF isoforms. Through analysis of crystal packing contacts and site-directed mutagenesis experiments we identified a conserved surface on the FHF core domain that mediates channel binding in vitro and in vivo. Mutations at this channel binding surface impaired the ability of FHFs to co-localize with Navs at the axon initial segment of hippocampal neurons. The mutations also disabled FHF modulation of voltage-dependent fast inactivation of sodium channels in neuronal cells. Based on our data, we propose that FHFs constitute auxiliary subunits for Navs.


Molecular and Cellular Neuroscience | 2009

FGF14 N-terminal splice variants differentially modulate Nav1.2 and Nav1.6-encoded sodium channels.

Fernanda Laezza; Angelika Lampert; Marie A. Kozel; Benjamin R. Gerber; Anthony M. Rush; Jeanne M. Nerbonne; Stephen G. Waxman; Sulayman D. Dib-Hajj; David M. Ornitz

The Intracellular Fibroblast Growth Factor (iFGF) subfamily includes four members (FGFs 11-14) of the structurally related FGF superfamily. Previous studies showed that the iFGFs interact directly with the pore-forming (alpha) subunits of voltage-gated sodium (Nav) channels and regulate the functional properties of sodium channel currents. Sequence heterogeneity among the iFGFs is thought to confer specificity to this regulation. Here, we demonstrate that the two N-terminal alternatively spliced FGF14 variants, FGF14-1a and FGF14-1b, differentially regulate currents produced by Nav1.2 and Nav1.6 channels. FGF14-1b, but not FGF14-1a, attenuates both Nav1.2 and Nav1.6 current densities. In contrast, co-expression of an FGF14 mutant, lacking the N-terminus, increased Nav1.6 current densities. In neurons, both FGF14-1a and FGF14-1b localized at the axonal initial segment, and deletion of the N-terminus abolished this localization. Thus, the FGF14 N-terminus is required for targeting and functional regulation of Nav channels, suggesting an important function for FGF14 alternative splicing in regulating neuronal excitability.


Molecular and Cellular Neuroscience | 2007

Impaired hippocampal synaptic transmission and plasticity in mice lacking fibroblast growth factor 14

Maolei Xiao; Lin Xu; Fernanda Laezza; Kelvin A. Yamada; Sheng Feng; David M. Ornitz

Humans with an autosomal dominant missense mutation in fibroblast growth factor 14 (FGF14) have impaired cognitive abilities and slowly progressive spinocerebellar ataxia. To explore the mechanisms that may account for this phenotype, we show that synaptic transmission at hippocampal Schaffer collateral-CA1 synapses and short- and long-term potentiation are impaired in Fgf14-/- mice, indicating abnormalities in synaptic plasticity. Examination of CA1 synapses in Fgf14-/- mice show a significant reduction in the number of synaptic vesicles docked at presynaptic active zones and a significant synaptic fatigue/depression during high/low-frequency stimulation. In addition, mEPSC frequency, but not amplitude, is decreased in hippocampal neurons derived from Fgf14-/- mice. Furthermore, expression of selective synaptic proteins in Fgf14-/- mice was decreased. These findings suggest a novel role for FGF14 in regulating synaptic plasticity via presynaptic mechanisms by affecting the mobilization, trafficking, or docking of synaptic vesicles to presynaptic active zones.


Journal of Biological Chemistry | 2013

The Fibroblast Growth Factor 14·Voltage-gated Sodium Channel Complex Is a New Target of Glycogen Synthase Kinase 3 (GSK3)

Alexander S. Shavkunov; Norelle C. Wildburger; Miroslav N. Nenov; Thomas F. James; Tetyana P. Buzhdygan; Neli I. Panova-Elektronova; Thomas A. Green; Ronald L. Veselenak; Nigel Bourne; Fernanda Laezza

Background: Fibroblast growth factor 14 (FGF14) binds to and regulates the voltage-gated Na+ (Nav) channel. Results: Inhibition of glycogen synthase kinase 3 (GSK3) modifies FGF14/Nav channel interaction, with effects on Na+ currents and subcellular distribution of the FGF14·Nav channel complex. Conclusion: The FGF14·Nav channel complex is a new target of GSK3. Significance: We provide evidence for modulation of Nav channels by GSK3 through FGF14. The FGF14 protein controls biophysical properties and subcellular distribution of neuronal voltage-gated Na+ (Nav) channels through direct binding to the channel C terminus. To gain insights into the dynamic regulation of this protein/protein interaction complex, we employed the split luciferase complementation assay to screen a small molecule library of kinase inhibitors against the FGF14·Nav1.6 channel complex and identified inhibitors of GSK3 as hits. Through a combination of a luminescence-based counter-screening, co-immunoprecipitation, patch clamp electrophysiology, and quantitative confocal immunofluorescence, we demonstrate that inhibition of GSK3 reduces the assembly of the FGF14·Nav channel complex, modifies FGF14-dependent regulation of Na+ currents, and induces dissociation and subcellular redistribution of the native FGF14·Nav channel complex in hippocampal neurons. These results further emphasize the role of FGF14 as a critical component of the Nav channel macromolecular complex, providing evidence for a novel GSK3-dependent signaling pathway that might control excitability through specific protein/protein interactions.


Journal of Neurochemistry | 2011

Dysregulated phosphorylation of Ca(2+) /calmodulin-dependent protein kinase II-α in the hippocampus of subjects with mild cognitive impairment and Alzheimer's disease.

Lindsay Reese; Fernanda Laezza; Randall L. Woltjer; Giulio Taglialatela

J. Neurochem. (2011) 119, 791–804.


Molecular and Cellular Neuroscience | 2007

KRIP6: A novel BTB/kelch protein regulating function of kainate receptors

Fernanda Laezza; Timothy J. Wilding; Sunitha M. Sequeira; Françoise Coussen; Xue Zhao Zhang; Rona Hill-Robinson; Christophe Mulle; James E. Huettner; Ann Marie Craig

Whereas many interacting proteins have been identified for AMPA and NMDA glutamate receptors, fewer are known to directly bind and regulate function of kainate receptors. Using a yeast two-hybrid screen for interacting partners of the C-terminal domain of GluR6a, we identified a novel neuronal protein of the BTB/kelch family, KRIP6. KRIP6 binds to the GluR6a C-terminal domain at a site distinct from the PDZ-binding motif and it co-immunoprecipitates with recombinant and endogenous GluR6. Co-expression of KRIP6 alters GluR6 mediated currents in a heterologous expression system reducing peak current amplitude and steady-state desensitization, without affecting surface levels of GluR6. Endogenous KRIP6 is widely expressed in brain and overexpression of KRIP6 reduces endogenous kainate receptor-mediated responses evoked in hippocampal neurons. Taken together, these results suggest that KRIP6 can directly regulate native kainate receptors and provide the first evidence for a BTB/kelch protein in direct functional regulation of a mammalian glutamate receptor.


The Journal of Neuroscience | 2014

Cognitive Enhancing Treatment with a PPARγ Agonist Normalizes Dentate Granule Cell Presynaptic Function in Tg2576 APP Mice

Miroslav N. Nenov; Fernanda Laezza; Sigmund J. Haidacher; Yingxin Zhao; Rovshan G. Sadygov; Jonathan M. Starkey; Heidi Spratt; Bruce A. Luxon; Kelly T. Dineley; Larry Denner

Hippocampal network hyperexcitability is considered an early indicator of Alzheimers disease (AD) memory impairment. Some AD mouse models exhibit similar network phenotypes. In this study we focused on dentate gyrus (DG) granule cell spontaneous and evoked properties in 9-month-old Tg2576 mice that model AD amyloidosis and cognitive deficits. Using whole-cell patch-clamp recordings, we found that Tg2576 DG granule cells exhibited spontaneous EPSCs that were higher in frequency but not amplitude compared with wild-type mice, suggesting hyperactivity of DG granule cells via a presynaptic mechanism. Further support of a presynaptic mechanism was revealed by increased I–O relationships and probability of release in Tg2576 DG granule cells. Since we and others have shown that activation of the peroxisome proliferator-activated receptor gamma (PPARγ) axis improves hippocampal cognition in mouse models for AD as well as benefitting memory performance in some humans with early AD, we investigated how PPARγ agonism affected synaptic activity in Tg2576 DG. We found that PPARγ agonism normalized the I–O relationship of evoked EPSCs, frequency of spontaneous EPSCs, and probability of release that, in turn, correlated with selective expression of DG proteins essential for presynaptic SNARE function that are altered in patients with AD. These findings provide evidence that DG principal cells may contribute to early AD hippocampal network hyperexcitability via a presynaptic mechanism, and that hippocampal cognitive enhancement via PPARγ activation occurs through regulation of presynaptic vesicular proteins critical for proper glutamatergic neurotransmitter release, synaptic transmission, and short-term plasticity.


Assay and Drug Development Technologies | 2012

Bioluminescence Methodology for the Detection of Protein–Protein Interactions Within the Voltage-Gated Sodium Channel Macromolecular Complex

Alexander S. Shavkunov; Neli Panova; Anesh Prasai; Ron Veselenak; Nigel Bourne; Svetla Stoilova-McPhie; Fernanda Laezza

Protein-protein interactions are critical molecular determinants of ion channel function and emerging targets for pharmacological interventions. Yet, current methodologies for the rapid detection of ion channel macromolecular complexes are still lacking. In this study we have adapted a split-luciferase complementation assay (LCA) for detecting the assembly of the voltage-gated Na+ (Nav) channel C-tail and the intracellular fibroblast growth factor 14 (FGF14), a functionally relevant component of the Nav channelosome that controls gating and targeting of Nav channels through direct interaction with the channel C-tail. In the LCA, two complementary N-terminus and C-terminus fragments of the firefly luciferase were fused, respectively, to a chimera of the CD4 transmembrane segment and the C-tail of Nav1.6 channel (CD4-Nav1.6-NLuc) or FGF14 (CLuc-FGF14). Co-expression of CLuc-FGF14 and CD4-Nav1.6-NLuc in live cells led to a robust assembly of the FGF14:Nav1.6 C-tail complex, which was attenuated by introducing single-point mutations at the predicted FGF14:Nav channel interface. To evaluate the dynamic regulation of the FGF14:Nav1.6 C-tail complex by signaling pathways, we investigated the effect of kinase inhibitors on the complex formation. Through a platform of counter screenings, we show that the p38/MAPK inhibitor, PD169316, and the IκB kinase inhibitor, BAY 11-7082, reduce the FGF14:Nav1.6 C-tail complementation, highlighting a potential role of the p38MAPK and the IκB/NFκB pathways in controlling neuronal excitability through protein-protein interactions. We envision the methodology presented here as a new valuable tool to allow functional evaluations of protein-channel complexes toward probe development and drug discovery targeting ion channels implicated in human disorders.

Collaboration


Dive into the Fernanda Laezza's collaboration.

Top Co-Authors

Avatar

Miroslav N. Nenov

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Syed R. Ali

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Aditya K. Singh

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Alexander S. Shavkunov

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Norelle C. Wildburger

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Thomas A. Green

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Neli Panova

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Federico Scala

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge