Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns

Abstract Background Fragile X syndrome (FXS), a neurodevelopmental disorder, is a leading monogenetic cause of intellectual disability and autism spectrum disorder. Notwithstanding the extensive studies using rodent and other pre-clinical models of FXS, which have provided detailed mechanistic insig...

Full description

Bibliographic Details
Main Authors: Shreya Das Sharma, Rakhi Pal, Bharath Kumar Reddy, Bhuvaneish T. Selvaraj, Nisha Raj, Krishna Kumar Samaga, Durga J. Srinivasan, Loren Ornelas, Dhruv Sareen, Matthew R. Livesey, Gary J. Bassell, Clive N. Svendsen, Peter C. Kind, Siddharthan Chandran, Sumantra Chattarji, David J. A. Wyllie
Format: Article
Language:English
Published: BMC 2020-06-01
Series:Molecular Autism
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13229-020-00351-4
id doaj-be1ddd0d597d4f079af05cef176f7f96
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Shreya Das Sharma
Rakhi Pal
Bharath Kumar Reddy
Bhuvaneish T. Selvaraj
Nisha Raj
Krishna Kumar Samaga
Durga J. Srinivasan
Loren Ornelas
Dhruv Sareen
Matthew R. Livesey
Gary J. Bassell
Clive N. Svendsen
Peter C. Kind
Siddharthan Chandran
Sumantra Chattarji
David J. A. Wyllie
spellingShingle Shreya Das Sharma
Rakhi Pal
Bharath Kumar Reddy
Bhuvaneish T. Selvaraj
Nisha Raj
Krishna Kumar Samaga
Durga J. Srinivasan
Loren Ornelas
Dhruv Sareen
Matthew R. Livesey
Gary J. Bassell
Clive N. Svendsen
Peter C. Kind
Siddharthan Chandran
Sumantra Chattarji
David J. A. Wyllie
Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
Molecular Autism
Fragile X syndrome
Disease-modelling
Electrophysiology
Action potential
author_facet Shreya Das Sharma
Rakhi Pal
Bharath Kumar Reddy
Bhuvaneish T. Selvaraj
Nisha Raj
Krishna Kumar Samaga
Durga J. Srinivasan
Loren Ornelas
Dhruv Sareen
Matthew R. Livesey
Gary J. Bassell
Clive N. Svendsen
Peter C. Kind
Siddharthan Chandran
Sumantra Chattarji
David J. A. Wyllie
author_sort Shreya Das Sharma
title Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
title_short Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
title_full Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
title_fullStr Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
title_full_unstemmed Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns
title_sort cortical neurons derived from human pluripotent stem cells lacking fmrp display altered spontaneous firing patterns
publisher BMC
series Molecular Autism
issn 2040-2392
publishDate 2020-06-01
description Abstract Background Fragile X syndrome (FXS), a neurodevelopmental disorder, is a leading monogenetic cause of intellectual disability and autism spectrum disorder. Notwithstanding the extensive studies using rodent and other pre-clinical models of FXS, which have provided detailed mechanistic insights into the pathophysiology of this disorder, it is only relatively recently that human stem cell-derived neurons have been employed as a model system to further our understanding of the pathophysiological events that may underlie FXS. Our study assesses the physiological properties of human pluripotent stem cell-derived cortical neurons lacking fragile X mental retardation protein (FMRP). Methods Electrophysiological whole-cell voltage- and current-clamp recordings were performed on two control and three FXS patient lines of human cortical neurons derived from induced pluripotent stem cells. In addition, we also describe the properties of an isogenic pair of lines in one of which FMR1 gene expression has been silenced. Results Neurons lacking FMRP displayed bursts of spontaneous action potential firing that were more frequent but shorter in duration compared to those recorded from neurons expressing FMRP. Inhibition of large conductance Ca2+-activated K+ currents and the persistent Na+ current in control neurons phenocopies action potential bursting observed in neurons lacking FMRP, while in neurons lacking FMRP pharmacological potentiation of voltage-dependent Na+ channels phenocopies action potential bursting observed in control neurons. Notwithstanding the changes in spontaneous action potential firing, we did not observe any differences in the intrinsic properties of neurons in any of the lines examined. Moreover, we did not detect any differences in the properties of miniature excitatory postsynaptic currents in any of the lines. Conclusions Pharmacological manipulations can alter the action potential burst profiles in both control and FMRP-null human cortical neurons, making them appear like their genetic counterpart. Our studies indicate that FMRP targets that have been found in rodent models of FXS are also potential targets in a human-based model system, and we suggest potential mechanisms by which activity is altered.
topic Fragile X syndrome
Disease-modelling
Electrophysiology
Action potential
url http://link.springer.com/article/10.1186/s13229-020-00351-4
work_keys_str_mv AT shreyadassharma corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT rakhipal corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT bharathkumarreddy corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT bhuvaneishtselvaraj corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT nisharaj corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT krishnakumarsamaga corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT durgajsrinivasan corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT lorenornelas corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT dhruvsareen corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT matthewrlivesey corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT garyjbassell corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT clivensvendsen corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT peterckind corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT siddharthanchandran corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT sumantrachattarji corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
AT davidjawyllie corticalneuronsderivedfromhumanpluripotentstemcellslackingfmrpdisplayalteredspontaneousfiringpatterns
_version_ 1724503632102031360
spelling doaj-be1ddd0d597d4f079af05cef176f7f962020-11-25T03:47:04ZengBMCMolecular Autism2040-23922020-06-0111111610.1186/s13229-020-00351-4Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patternsShreya Das Sharma0Rakhi Pal1Bharath Kumar Reddy2Bhuvaneish T. Selvaraj3Nisha Raj4Krishna Kumar Samaga5Durga J. Srinivasan6Loren Ornelas7Dhruv Sareen8Matthew R. Livesey9Gary J. Bassell10Clive N. Svendsen11Peter C. Kind12Siddharthan Chandran13Sumantra Chattarji14David J. A. Wyllie15Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Clinical Brain Sciences, University of EdinburghDepartment of Cell Biology, Emory University School of MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineThe Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical CenterThe Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical CenterCentre for Discovery Brain Sciences, University of EdinburghDepartment of Cell Biology, Emory University School of MedicineThe Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical CenterCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative MedicineAbstract Background Fragile X syndrome (FXS), a neurodevelopmental disorder, is a leading monogenetic cause of intellectual disability and autism spectrum disorder. Notwithstanding the extensive studies using rodent and other pre-clinical models of FXS, which have provided detailed mechanistic insights into the pathophysiology of this disorder, it is only relatively recently that human stem cell-derived neurons have been employed as a model system to further our understanding of the pathophysiological events that may underlie FXS. Our study assesses the physiological properties of human pluripotent stem cell-derived cortical neurons lacking fragile X mental retardation protein (FMRP). Methods Electrophysiological whole-cell voltage- and current-clamp recordings were performed on two control and three FXS patient lines of human cortical neurons derived from induced pluripotent stem cells. In addition, we also describe the properties of an isogenic pair of lines in one of which FMR1 gene expression has been silenced. Results Neurons lacking FMRP displayed bursts of spontaneous action potential firing that were more frequent but shorter in duration compared to those recorded from neurons expressing FMRP. Inhibition of large conductance Ca2+-activated K+ currents and the persistent Na+ current in control neurons phenocopies action potential bursting observed in neurons lacking FMRP, while in neurons lacking FMRP pharmacological potentiation of voltage-dependent Na+ channels phenocopies action potential bursting observed in control neurons. Notwithstanding the changes in spontaneous action potential firing, we did not observe any differences in the intrinsic properties of neurons in any of the lines examined. Moreover, we did not detect any differences in the properties of miniature excitatory postsynaptic currents in any of the lines. Conclusions Pharmacological manipulations can alter the action potential burst profiles in both control and FMRP-null human cortical neurons, making them appear like their genetic counterpart. Our studies indicate that FMRP targets that have been found in rodent models of FXS are also potential targets in a human-based model system, and we suggest potential mechanisms by which activity is altered.http://link.springer.com/article/10.1186/s13229-020-00351-4Fragile X syndromeDisease-modellingElectrophysiologyAction potential