1
|
Hussain A, Razak KA. Perineuronal net degradation causes a delayed change in resting and sound evoked responses in the mouse auditory cortex. Neuroscience 2025; 577:252-263. [PMID: 40389125 DOI: 10.1016/j.neuroscience.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Perineuronal nets (PNNs) are extracellular matrix assemblies that preferentially cover parvalbumin-expressing (PV+) interneurons in the neocortex. PV+ cells and PNNs are impaired in a variety of neurodevelopmental disorders including Fragile X Syndrome and schizophrenia. In both of these disorders, electroencephalograph (EEG) recordings show similar phenotypes, including elevated resting gamma band power and reduced temporal fidelity in the 40 Hz auditory steady state response (ASSR). Whether there is a causal link between PNN integrity and EEG abnormalities remains unclear. We tested this link by recording EEG responses in the auditory cortex (AC) in wildtype mice in which PNNs were enzymatically degraded (Chondroitinase ABC or ChABC). EEGs were recorded at two different time points (4- or 14-days post injection, cross-sectional design). In comparison to saline control, ChABC injected mice showed a ∼50 % reduction in PNN density after 4-days. However, there was no difference in resting EEG power spectral density, auditory event-related potential amplitudes or ASSR temporal fidelity between saline and ChABC mice. At the 14-day time point, there was a recovery of PNN density in the AC. Interestingly, EEG responses were abnormal at this time point, with elevated gamma band activity and reduced ASSR temporal fidelity. Thus, the electrophysiological consequences of PNN loss are not seen acutely, but over a delayed time course, suggesting abnormal plasticity after a circuit perturbation. Taken together, these data indicate acute shaping of auditory cortical responses is less dependent on PNNs, but long-term stability of responses following a circuit perturbation depends on the integrity of PNNs.
Collapse
Affiliation(s)
- A Hussain
- Graduate Neuroscience Program, University of California, Riverside, USA
| | - K A Razak
- Graduate Neuroscience Program, University of California, Riverside, USA; Department of Psychology, University of California, Riverside, USA.
| |
Collapse
|
2
|
Norman AO, Farooq N, Sahni A, Tapia K, Breiner D, Razak KA, Ethell IM. Differential effects of sound repetition rate on auditory cortex development and behavior in fragile X syndrome mouse model. Exp Neurol 2025; 387:115184. [PMID: 39961384 DOI: 10.1016/j.expneurol.2025.115184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Fragile X syndrome (FXS) is a leading genetic form of autism and intellectual disability that is associated with a loss-of-function mutation in the Fragile X messenger ribonucleoprotein 1 (Fmr1) gene. The Fmr1 knockout (KO) mouse model displays many aspects of FXS-related phenotypes and is used to study FXS pathophysiology. Sensory manipulations, such as sound exposure, are considered as a non-invasive approach to alleviate FXS phenotypes. However, it is unclear what specific sound attributes may have beneficial effects. In this study, we examined the effects of sound repetition rate on auditory cortex development and FXS-associated behaviors in a mouse model of FXS. KO and wild-type (WT) male littermates were exposed to 14 kHz pure tone trains with 1 Hz or 5 Hz repetition rates during postnatal day (P)9-P21 developmental period. We analyzed the effects of developmental sound exposure on PV cell development, cortical activity and exploratory behaviors in sound-exposed WT and KO mice. We found that parvalbumin (PV) cell density was lower in the auditory cortex (AuC) of KO compared to WT mice raised in sound-attenuated environment, but was increased following the exposure to both 1 Hz and 5 Hz sound trains. However, PV protein levels were upregulated only in AuC of 5 Hz rate exposed KO mice. Interestingly, analysis of baseline cortical activity using electroencephalography (EEG) recordings showed that sound attenuation or exposure to sound trains with 5 Hz, but not 1 Hz, repetition rates corrected enhanced resting state gamma power in AuC of KO mice to WT levels. In addition, sound attenuation and exposure to 5 Hz showed some beneficial effects on the synchronization to frequency-modulated chirp in the frontal cortex (FC) of both WT and KO mice. Analysis of event-related potentials (ERP) in response to broadband sound showed increased ongoing responses and decreased habituation to noise stimuli in the AuC and FC of naive KO mice. While sound-attenuation and exposure to 5 Hz showed no significant effects on the power of onset and ongoing responses, exposure to 1 Hz further enhanced ongoing responses and decreased habituation to sound in both WT and KO mice. Finally, developmental exposure to sound trains with 5 Hz, but not 1 Hz, repetition rates normalized exploratory behaviors and improved social novelty preference but not hyperactivity in KO mice. Summarizing, our results show that developmental exposure of mice to sound trains with 5 Hz, but not 1 Hz, repetition rate had beneficial effects on PV cell development, overall cortical activity and behaviors in KO mice. While sound attenuation alone normalized some EEG phenotypes, it did not improve PV development or behaviors. These findings may have a significant impact on developing new approaches to alleviate FXS phenotypes and open possibilities for a combination of sound exposure with drug treatment which may offer highly novel therapeutic approaches.
Collapse
Affiliation(s)
- A O Norman
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - N Farooq
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - A Sahni
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - K Tapia
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - D Breiner
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - K A Razak
- Graduate Neuroscience Program, University of California Riverside, Riverside, CA, USA; Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - I M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Graduate Neuroscience Program, University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
3
|
van der Lei MB, Kooy RF. From Discovery to Innovative Translational Approaches in 80 Years of Fragile X Syndrome Research. Biomedicines 2025; 13:805. [PMID: 40299377 PMCID: PMC12024745 DOI: 10.3390/biomedicines13040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and a major genetic contributor to autism spectrum disorder. It is caused by a CGG trinucleotide repeat expansion in the FMR1 gene, resulting in gene silencing and the loss of FMRP, an RNA-binding protein essential for synaptic plasticity. This review covers over 80 years of FXS research, highlighting key milestones, clinical features, genetic and molecular mechanisms, the FXS mouse model, disrupted molecular pathways, and current therapeutic strategies. Additionally, we discuss recent advances including AI-driven combination therapies, CRISPR-based gene editing, and antisense oligonucleotides (ASOs) therapies. Despite these scientific breakthroughs, translating preclinical findings into effective clinical treatments remains challenging. Clinical trials have faced several difficulties, including patient heterogeneity, inconsistent outcome measures, and variable therapeutic responses. Standardized preclinical testing protocols and refined clinical trial designs are required to overcome these challenges. The development of FXS-specific biomarkers could also improve the precision of treatment assessments. Ultimately, future therapies will need to combine pharmacological and behavioral interventions tailored to individual needs. While significant challenges remain, ongoing research continues to offer hope for transformative breakthroughs that could significantly improve the quality of life for individuals with FXS and their families.
Collapse
Affiliation(s)
| | - R. Frank Kooy
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium;
| |
Collapse
|
4
|
Gauthier DW, James N, Auerbach BD. Altered auditory feature discrimination in a rat model of Fragile X Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638956. [PMID: 40027738 PMCID: PMC11870463 DOI: 10.1101/2025.02.18.638956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Atypical sensory processing, particularly in the auditory domain, is one of the most common and quality-of-life affecting symptoms seen in autism spectrum disorders (ASD). Fragile X Syndrome (FXS) is the leading inherited cause of ASD and a majority of FXS individuals present with auditory processing alterations. While auditory hypersensitivity is a common phenotype observed in FXS and Fmr1 KO rodent models, it is important to consider other auditory coding impairments that could contribute to sound processing difficulties and disrupted language comprehension in FXS. We have shown previously that a Fmr1 knockout (KO) rat model of FXS exhibits heightened sound sensitivity that coincided with abnormal perceptual integration of sound bandwidth, indicative of altered spectral processing. Frequency discrimination is a fundamental aspect of sound encoding that is important for a range of auditory processes, such as source segregation and speech comprehension, and disrupted frequency coding could thus contribute to a range of auditory issues in FXS and ASD. Here we explicitly characterized spectral processing deficits in male Fmr1 KO rats using an operant conditioning tone discrimination assay and in vivo electrophysiology recordings from the auditory cortex and inferior colliculus. We found that Fmr1 KO rats exhibited poorer frequency resolution, which corresponded with neuronal hyperactivity and broader frequency tuning in auditory cortical but not collicular neurons. Using an experimentally informed population model, we show that these cortical physiological differences can recapitulate the observed behavior discrimination deficits, with decoder performance being tightly linked to differences in cortical tuning width and signal-to-noise ratios. These findings suggest that cortical hyperexcitability may account for a range of auditory behavioral phenotypes in FXS, providing a potential locus for development of novel biomarkers and treatment strategies that could extend to other forms of ASD.
Collapse
Affiliation(s)
- D. Walker Gauthier
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois Urbana-Champaign Urbana, Illinois, United States
- Neuroscience Program, University of Illinois Urbana-Champaign Urbana, Illinois, United States
| | - Noelle James
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois Urbana-Champaign Urbana, Illinois, United States
| | - Benjamin D. Auerbach
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois Urbana-Champaign Urbana, Illinois, United States
- Neuroscience Program, University of Illinois Urbana-Champaign Urbana, Illinois, United States
| |
Collapse
|
5
|
Lu C, Linden JF. Auditory evoked-potential abnormalities in a mouse model of 22q11.2 Deletion Syndrome and their interactions with hearing impairment. Transl Psychiatry 2025; 15:4. [PMID: 39779687 PMCID: PMC11711659 DOI: 10.1038/s41398-024-03218-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/02/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
The 22q11.2 deletion is a risk factor for multiple psychiatric disorders including schizophrenia and also increases vulnerability to middle-ear problems that can cause hearing impairment. Up to 60% of deletion carriers experience hearing impairment and ~30% develop schizophrenia in adulthood. It is not known if these risks interact. Here we used the Df1/+ mouse model of the 22q11.2 deletion to investigate how hearing impairment might interact with increased genetic vulnerability to psychiatric disease to affect brain function. We measured brain function using cortical auditory evoked potentials (AEPs), which are commonly measured non-invasively in humans. After identifying one of the simplest and best-validated methods for AEP measurement in mice from the diversity of previous approaches, we measured peripheral hearing sensitivity and cortical AEPs in Df1/+ mice and their WT littermates. We exploited large inter-individual variation in hearing ability among Df1/+ mice to distinguish effects of genetic background from effects of hearing impairment. Central auditory gain and adaptation were quantified by comparing brainstem activity and cortical AEPs and by analyzing the growth of cortical AEPs with increasing sound level or inter-tone interval duration. We found that level-dependent AEP growth was abnormally large in Df1/+ mice regardless of hearing impairment, but other AEP measures of central auditory gain and adaptation depended on both genotype and hearing phenotype. Our results demonstrate the relevance of comorbid hearing loss to auditory brain dysfunction in 22q11.2DS and also identify potential biomarkers for psychiatric disease that are robust to hearing impairment.
Collapse
Affiliation(s)
- Chen Lu
- Ear Institute, University College London, London, UK
| | - Jennifer F Linden
- Ear Institute, University College London, London, UK.
- Department of Neuroscience, Physiology, & Pharmacology, University College London, London, UK.
| |
Collapse
|
6
|
Tao X, Croom K, Newman-Tancredi A, Varney M, Razak KA. Acute administration of NLX-101, a Serotonin 1A receptor agonist, improves auditory temporal processing during development in a mouse model of Fragile X Syndrome. J Neurodev Disord 2025; 17:1. [PMID: 39754065 PMCID: PMC11697955 DOI: 10.1186/s11689-024-09587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading known genetic cause of intellectual disability and autism spectrum disorders (ASD)-associated behaviors. A consistent and debilitating phenotype of FXS is auditory hypersensitivity that may lead to delayed language and high anxiety. Consistent with findings in FXS human studies, the mouse model of FXS, the Fmr1 knock out (KO) mouse, shows auditory hypersensitivity and temporal processing deficits. In electroencephalograph (EEG) recordings from humans and mice, these deficits manifest as increased N1 amplitudes in event-related potentials (ERP), increased gamma band single trial power (STP) and reduced phase locking to rapid temporal modulations of sound. In our previous study, we found that administration of the selective serotonin-1 A (5-HT1A)receptor biased agonist, NLX-101, protected Fmr1 KO mice from auditory hypersensitivity-associated seizures. Here we tested the hypothesis that NLX-101 will normalize EEG phenotypes in developing Fmr1 KO mice. METHODS To test this hypothesis, we examined the effect of NLX-101 on EEG phenotypes in male and female wildtype (WT) and Fmr1 KO mice. Using epidural electrodes, we recorded auditory event related potentials (ERP) and auditory temporal processing with a gap-in-noise auditory steady state response (ASSR) paradigm at two ages, postnatal (P) 21 and 30 days, from both auditory and frontal cortices of awake, freely moving mice, following NLX-101 (at 1.8 mg/kg i.p.) or saline administration. RESULTS Saline-injected Fmr1 KO mice showed increased N1 amplitudes, increased STP and reduced phase locking to auditory gap-in-noise stimuli versus wild-type mice, reproducing previously published EEG phenotypes. An acute injection of NLX-101 did not alter ERP amplitudes at either P21 or P30, but significantly reduces STP at P30. Inter-trial phase clustering was significantly increased in both age groups with NLX-101, indicating improved temporal processing. The differential effects of serotonin modulation on ERP, background power and temporal processing suggest different developmental mechanisms leading to these phenotypes. CONCLUSIONS These results suggest that NLX-101 could constitute a promising treatment option for targeting post-synaptic 5-HT1A receptors to improve auditory temporal processing, which in turn may improve speech and language function in FXS.
Collapse
Affiliation(s)
- Xin Tao
- Graduate Neuroscience Program, University of California, Riverside, CA, USA
| | - Katilynne Croom
- Graduate Neuroscience Program, University of California, Riverside, CA, USA
| | | | | | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, CA, USA.
- Department of Psychology, University of California, 900 University Avenue, Riverside, CA, 92521, USA.
| |
Collapse
|
7
|
Kokash J, Rumschlag JA, Razak KA. Cortical region-specific recovery of auditory temporal processing following noise-induced hearing loss. Neuroscience 2024; 560:143-157. [PMID: 39284433 DOI: 10.1016/j.neuroscience.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024]
Abstract
Noise-induced hearing loss (NIHL) studies have focused on the lemniscal auditory pathway, but little is known about how NIHL impacts different cortical regions. Here we compared response recovery trajectories in the auditory and frontal cortices (AC, FC) of mice following NIHL. We recorded EEG responses from awake mice (male n = 15, female n = 14) before and following NIHL (longitudinal design) to quantify event related potentials and gap-in-noise temporal processing. Hearing loss was verified by measuring the auditory brainstem response (ABR) before and at 1-, 10-, 23-, and 45-days after noise-exposure. Resting EEG, event related potentials (ERP) and auditory steady state responses (ASSR) were recorded at the same time-points after NIHL. The inter-trial phase coherence (ITPC) of the ASSR was measured to quantify the ability of AC and FC to synchronize responses to short gaps embedded in noise. Despite the absence of click-evoked ABRs up to 90 dB SPL and up to 45-days post-exposure, ERPs from the AC and FC showed full recovery in ∼ 50 % of the mice to pre-NIHL levels in both AC and FC. The ASSR ITPC was reduced following NIHL in AC and FC in all the mice on day 1 after NIHL. The AC showed full recovery of ITPC over 45-days. Despite ERP amplitude recovery, the FC does not show recovery of ASSR ITPC. These results indicate post-NIHL plasticity with similar response amplitude recovery across AC and FC, but cortical region-specific trajectories in temporal processing recovery.
Collapse
Affiliation(s)
- J Kokash
- Graduate Neuroscience Program, University of California, Riverside, United States
| | - J A Rumschlag
- Graduate Neuroscience Program, University of California, Riverside, United States
| | - K A Razak
- Graduate Neuroscience Program, University of California, Riverside, United States; Department of Psychology, University of California, Riverside, United States.
| |
Collapse
|
8
|
Wilde M, Ghanbari A, Mancienne T, Moran A, Poulsen RE, Constantin L, Lee C, Scholz LA, Arnold J, Qin W, Karle TJ, Petrou S, Favre-Bulle I, Hoffman EJ, Scott EK. Brain-wide circuitry underlying altered auditory habituation in zebrafish models of autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611137. [PMID: 39282371 PMCID: PMC11398315 DOI: 10.1101/2024.09.04.611137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Auditory processing is widely understood to occur differently in autism, though the patterns of brain activity underlying these differences are not well understood. The diversity of autism also means brain-wide networks may change in various ways to produce similar behavioral outputs. We used larval zebrafish to investigate auditory habituation in four genetic lines relevant to autism: fmr1, mecp2, scn1lab and cntnap2. In free-swimming behavioral tests, we found each line had a unique profile of auditory hypersensitivity and/or delayed habituation. Combining the optical transparency of larval zebrafish with genetically encoded calcium indicators and light-sheet microscopy, we then observed brain-wide activity at cellular resolution during auditory habituation. As with behavior, each line showed unique alterations in brain-wide spontaneous activity, auditory processing, and adaptation in response to repetitive acoustic stimuli. We also observed commonalities in activity across our genetic lines that indicate shared circuit changes underlying certain aspects of their behavioral phenotypes. These were predominantly in regions involved in sensory integration and sensorimotor gating rather than primary auditory areas. Overlapping phenotypes include differences in the activity and functional connectivity of the telencephalon, thalamus, dopaminergic regions, and the locus coeruleus, and excitatory/inhibitory imbalance in the cerebellum. Unique phenotypes include loss of activity in the habenula in scn1lab, increased activity in auditory regions in fmr1, and differences in network activity over time in mecp2 and cntnap2. Comparing these distinct but overlapping brain-wide auditory networks furthers our understanding of how diverse genetic factors can produce similar behavioral effects through a range of circuit- and network-scale mechanisms.
Collapse
Affiliation(s)
- Maya Wilde
- Queensland Brain Institute, University of Queensland, QLD, Australia
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Anahita Ghanbari
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Tessa Mancienne
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Ailís Moran
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Rebecca E. Poulsen
- Department of Linguistics, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, Australia
| | - Lena Constantin
- Queensland Brain Institute, University of Queensland, QLD, Australia
| | - Conrad Lee
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Leandro Aluisio Scholz
- Queensland Brain Institute, University of Queensland, QLD, Australia
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Joshua Arnold
- Queensland Brain Institute, University of Queensland, QLD, Australia
| | - Wei Qin
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Timothy J. Karle
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Itia Favre-Bulle
- Queensland Brain Institute, University of Queensland, QLD, Australia
| | - Ellen J. Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Ethan K. Scott
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| |
Collapse
|
9
|
Jonak CR, Assad SA, Garcia TA, Sandhu MS, Rumschlag JA, Razak KA, Binder DK. Phenotypic analysis of multielectrode array EEG biomarkers in developing and adult male Fmr1 KO mice. Neurobiol Dis 2024; 195:106496. [PMID: 38582333 DOI: 10.1016/j.nbd.2024.106496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent electroencephalographic (EEG) studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced phase locking of sound-evoked gamma oscillations. Similar EEG phenotypes are present in mouse models of FXS, but very little is known about the development of such abnormal responses. In the current study, we employed a 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in male P21 and P91 WT and Fmr1 KO mice. This led to several novel findings. First, P91, but not P21, Fmr1 KO mice have significantly increased resting EEG power in the low- and high-gamma frequency bands. Second, both P21 and P91 Fmr1 KO mice have markedly attenuated inter-trial phase coherence (ITPC) to spectrotemporally dynamic auditory stimuli as well as to 40 Hz and 80 Hz auditory steady-state response (ASSR) stimuli. This suggests abnormal temporal processing from early development that may lead to abnormal speech and language function in FXS. Third, we found hemispheric asymmetry of fast temporal processing in the mouse auditory cortex in WT but not Fmr1 KO mice. Together, these findings define a set of EEG phenotypes in young and adult mice that can serve as translational targets for genetic and pharmacological manipulation in phenotypic rescue studies.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Samantha A Assad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Terese A Garcia
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Manbir S Sandhu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Jeffrey A Rumschlag
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, United States of America
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, CA, United States of America; Department of Psychology, University of California, Riverside, CA, United States of America
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America; Neuroscience Graduate Program, University of California, Riverside, CA, United States of America.
| |
Collapse
|
10
|
Wadle SL, Ritter TC, Wadle TTX, Hirtz JJ. Topography and Ensemble Activity in the Auditory Cortex of a Mouse Model of Fragile X Syndrome. eNeuro 2024; 11:ENEURO.0396-23.2024. [PMID: 38627066 PMCID: PMC11097631 DOI: 10.1523/eneuro.0396-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/11/2024] [Accepted: 04/01/2024] [Indexed: 05/18/2024] Open
Abstract
Autism spectrum disorder (ASD) is often associated with social communication impairments and specific sound processing deficits, for example, problems in following speech in noisy environments. To investigate underlying neuronal processing defects located in the auditory cortex (AC), we performed two-photon Ca2+ imaging in FMR1 (fragile X messenger ribonucleoprotein 1) knock-out (KO) mice, a model for fragile X syndrome (FXS), the most common cause of hereditary ASD in humans. For primary AC (A1) and the anterior auditory field (AAF), topographic frequency representation was less ordered compared with control animals. We additionally analyzed ensemble AC activity in response to various sounds and found subfield-specific differences. In A1, ensemble correlations were lower in general, while in secondary AC (A2), correlations were higher in response to complex sounds, but not to pure tones. Furthermore, sound specificity of ensemble activity was decreased in AAF. Repeating these experiments 1 week later revealed no major differences regarding representational drift. Nevertheless, we found subfield- and genotype-specific changes in ensemble correlation values between the two times points, hinting at alterations in network stability in FMR1 KO mice. These detailed insights into AC network activity and topography in FMR1 KO mice add to the understanding of auditory processing defects in FXS.
Collapse
Affiliation(s)
- Simon L Wadle
- Physiology of Neuronal Networks, Department of Biology, RPTU University of Kaiserslautern-Landau, Kaiserslautern D-67663, Germany
| | - Tamara C Ritter
- Physiology of Neuronal Networks, Department of Biology, RPTU University of Kaiserslautern-Landau, Kaiserslautern D-67663, Germany
| | - Tatjana T X Wadle
- Physiology of Neuronal Networks, Department of Biology, RPTU University of Kaiserslautern-Landau, Kaiserslautern D-67663, Germany
| | - Jan J Hirtz
- Physiology of Neuronal Networks, Department of Biology, RPTU University of Kaiserslautern-Landau, Kaiserslautern D-67663, Germany
| |
Collapse
|
11
|
Liu R, Pedapati EV, Schmitt LM, Shaffer RC, Smith EG, Dominick KC, DeStefano LA, Westerkamp G, Horn P, Sweeney JA, Erickson CA. Reliability of resting-state electrophysiology in fragile X syndrome. Biomark Neuropsychiatry 2023; 9:100070. [PMID: 38817342 PMCID: PMC11138258 DOI: 10.1016/j.bionps.2023.100070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
Objective Fragile X Syndrome (FXS) is the leading monogenic cause of intellectual disability and autism spectrum disorder. Currently, there are no established biomarkers for predicting and monitoring drug effects in FXS, and no approved therapies are available. Previous studies have shown electrophysiological changes in the brain using electroencephalography (EEG) in individuals with FXS and animal models. These changes may be influenced by drug therapies. In this study, we aimed to assess the reliability of resting-state EEG measures in individuals with FXS, which could potentially serve as a biomarker for drug discovery. Methods We collected resting-state EEG data from 35 individuals with FXS participating in placebo-controlled clinical trials (23 males, 12 females; visit age mean+/-std 25.6 +/-8.3). The data were analyzed for various spectral features using intraclass correlation analysis to evaluate test-retest reliability. The intervals between EEG recordings ranged from same-day measurements to up to six weeks apart. Results Our results showed high reliability for most spectral features, with same-day reliability exceeding 0.8. Features of interest demonstrated ICC values of 0.60 or above at longer intervals. Among the features, alpha band relative power exhibited the highest reliability. Conclusion These findings indicate that resting-state EEG can provide consistent and reproducible measures of brain activity in individuals with FXS. This supports the potential use of EEG as an objective biomarker for evaluating the effects of new drugs in FXS. Significance The reliable measurements obtained from power spectrum-based resting-state EEG make it a promising tool for assessing the impact of small molecule drugs in FXS.
Collapse
Affiliation(s)
- Rui Liu
- Cincinnati Children’s Hospital Medical Center, United States
| | - Ernest V. Pedapati
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | - Lauren M. Schmitt
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | - Rebecca C. Shaffer
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | - Elizabeth G. Smith
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | - Kelli C. Dominick
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | | | | | - Paul Horn
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| | | | - Craig A. Erickson
- Cincinnati Children’s Hospital Medical Center, United States
- University of Cincinnati, United States
| |
Collapse
|
12
|
Monday HR, Wang HC, Feldman DE. Circuit-level theories for sensory dysfunction in autism: convergence across mouse models. Front Neurol 2023; 14:1254297. [PMID: 37745660 PMCID: PMC10513044 DOI: 10.3389/fneur.2023.1254297] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Individuals with autism spectrum disorder (ASD) exhibit a diverse range of behavioral features and genetic backgrounds, but whether different genetic forms of autism involve convergent pathophysiology of brain function is unknown. Here, we analyze evidence for convergent deficits in neural circuit function across multiple transgenic mouse models of ASD. We focus on sensory areas of neocortex, where circuit differences may underlie atypical sensory processing, a central feature of autism. Many distinct circuit-level theories for ASD have been proposed, including increased excitation-inhibition (E-I) ratio and hyperexcitability, hypofunction of parvalbumin (PV) interneuron circuits, impaired homeostatic plasticity, degraded sensory coding, and others. We review these theories and assess the degree of convergence across ASD mouse models for each. Behaviorally, our analysis reveals that innate sensory detection behavior is heightened and sensory discrimination behavior is impaired across many ASD models. Neurophysiologically, PV hypofunction and increased E-I ratio are prevalent but only rarely generate hyperexcitability and excess spiking. Instead, sensory tuning and other aspects of neural coding are commonly degraded and may explain impaired discrimination behavior. Two distinct phenotypic clusters with opposing neural circuit signatures are evident across mouse models. Such clustering could suggest physiological subtypes of autism, which may facilitate the development of tailored therapeutic approaches.
Collapse
Affiliation(s)
- Hannah R. Monday
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | | | - Daniel E. Feldman
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
13
|
Croom K, Rumschlag JA, Erickson MA, Binder DK, Razak KA. Developmental delays in cortical auditory temporal processing in a mouse model of Fragile X syndrome. J Neurodev Disord 2023; 15:23. [PMID: 37516865 PMCID: PMC10386252 DOI: 10.1186/s11689-023-09496-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) encompass a wide array of debilitating symptoms, including sensory dysfunction and delayed language development. Auditory temporal processing is crucial for speech perception and language development. Abnormal development of temporal processing may account for the language impairments associated with ASD. Very little is known about the development of temporal processing in any animal model of ASD. METHODS In the current study, we quantify auditory temporal processing throughout development in the Fmr1 knock-out (KO) mouse model of Fragile X Syndrome (FXS), a leading genetic cause of intellectual disability and ASD-associated behaviors. Using epidural electrodes in awake and freely moving wildtype (WT) and KO mice, we recorded auditory event related potentials (ERP) and auditory temporal processing with a gap-in-noise auditory steady state response (gap-ASSR) paradigm. Mice were recorded at three different ages in a cross sectional design: postnatal (p)21, p30 and p60. Recordings were obtained from both auditory and frontal cortices. The gap-ASSR requires underlying neural generators to synchronize responses to gaps of different widths embedded in noise, providing an objective measure of temporal processing across genotypes and age groups. RESULTS We present evidence that the frontal, but not auditory, cortex shows significant temporal processing deficits at p21 and p30, with poor ability to phase lock to rapid gaps in noise. Temporal processing was similar in both genotypes in adult mice. ERP amplitudes were larger in Fmr1 KO mice in both auditory and frontal cortex, consistent with ERP data in humans with FXS. CONCLUSIONS These data indicate cortical region-specific delays in temporal processing development in Fmr1 KO mice. Developmental delays in the ability of frontal cortex to follow rapid changes in sounds may shape language delays in FXS, and more broadly in ASD.
Collapse
Affiliation(s)
- Katilynne Croom
- Graduate Neuroscience Program, University of California, Riverside, USA
| | - Jeffrey A Rumschlag
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, USA
| | | | - Devin K Binder
- Graduate Neuroscience Program, University of California, Riverside, USA
- Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, USA.
- Department of Psychology, University of California, Riverside, USA.
| |
Collapse
|
14
|
Ethridge LE, Auerbach BD, Contractor A, Ethell IM, McCullagh EA, Pedapati EV. Editorial: Neural markers of sensory processing in development. Front Integr Neurosci 2023; 17:1256437. [PMID: 37547460 PMCID: PMC10401584 DOI: 10.3389/fnint.2023.1256437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023] Open
Affiliation(s)
- Lauren E. Ethridge
- Department of Psychology, University of Oklahoma, Norman, OK, United States
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Benjamin D. Auerbach
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Anis Contractor
- Department of Neuroscience, Psychiatry and Behavioral Sciences, and Neurobiology, Northwestern University, Evanston, IL, United States
| | - Iryna M. Ethell
- Department of Biomedical Sciences, University of California, Riverside, Riverside, CA, United States
| | - Elizabeth A. McCullagh
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| | - Ernest V. Pedapati
- Department of Psychiatry and Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Division of Child and Adolescent Psychiatry, and Child Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
15
|
Bigras C, Villatte B, Duda V, Hébert S. The electrophysiological markers of hyperacusis: a scoping review. Int J Audiol 2023; 62:489-499. [PMID: 35549972 DOI: 10.1080/14992027.2022.2070083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/05/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Hyperacusis is known as a reduced tolerance to sounds perceived as normal to the majority of the population. There is currently no agreed definition, diagnostic tool, or objective measure of its occurrence. The purpose of this review is to catalogue the research to date on the use of auditory evoked potentials (AEP) to assess hyperacusis. DESIGN A step-by-step methodology was conducted following guidelines. Four databases were searched. A total of 3343 papers were identified. A final yield of 35 articles were retained for analysis. RESULTS The analysis identified four types of aetiologies to describe the hyperacusic population in AEP studies; developmental disorders (n = 19), neurological disorders (n = 3), induced hearing damage (n = 8) and idiopathic aetiology (n = 5). Electrophysiological measures were of short (n = 16), middle (n = 13) and long (n = 19) latencies, believed to reflect the activity of the ascending and descending pathways of the auditory system from periphery to cortex. CONCLUSIONS The results of this review revealed the potential use of electrophysiological measures for further understanding the mechanisms of hyperacusis. However, according to the disparity of concepts to define hyperacusis, definitions and populations need to be clarified before biomarkers specific to hyperacusis can be identified.
Collapse
Affiliation(s)
- Charlotte Bigras
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada
- Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| | - Bérangère Villatte
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada
- Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| | - Victoria Duda
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada
- Centre de recherche interdisciplinaire en réadaptation (CRIR), Montreal, Canada
| | - Sylvie Hébert
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada
- Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| |
Collapse
|
16
|
Ren B, Burkovetskaya M, Jung Y, Bergdolt L, Totusek S, Martinez-Cerdeno V, Stauch K, Korade Z, Dunaevsky A. Dysregulated cholesterol metabolism, aberrant excitability and altered cell cycle of astrocytes in fragile X syndrome. Glia 2023; 71:1176-1196. [PMID: 36594399 PMCID: PMC10023374 DOI: 10.1002/glia.24331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023]
Abstract
Fragile X syndrome (FXS), the most prevalent heritable form of intellectual disability, is caused by the transcriptional silencing of the FMR1 gene. While neuronal contribution to FXS has been extensively studied in both animal and human-based models of FXS, the roles of astrocytes, a type of glial cells in the brain, are largely unknown. Here, we generated a human-based FXS model via differentiation of astrocytes from human-induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) and characterized their development, function, and proteomic profiles. We identified shortened cell cycle, enhanced Ca2+ signaling, impaired sterol biosynthesis, and pervasive alterations in the proteome of FXS astrocytes. Our work identified astrocytic impairments that could contribute to the pathogenesis of FXS and highlight astrocytes as a novel therapeutic target for FXS treatment.
Collapse
Affiliation(s)
- Baiyan Ren
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Maria Burkovetskaya
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yoosun Jung
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Lara Bergdolt
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steven Totusek
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Veronica Martinez-Cerdeno
- Department of Pathology and Laboratory Medicine, MIND Institute, and Institute for Pediatric Regenerative Medicine at UC Davis School of Medicine, and Shriners Hospitals for Children of Northern California, Sacramento, California, USA
| | - Kelly Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Zeljka Korade
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pediatrics, CHRI, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anna Dunaevsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
17
|
Not Mismatch Negativity, but Modulation of Sensory N1 is Measured - Comment to Chen-Engerer et al. Neuroscience 2023; 512:133-134. [PMID: 36549604 DOI: 10.1016/j.neuroscience.2022.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
|
18
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
19
|
Bertocchi I, Cambiaghi M, Hasan MT. Advances toward precision therapeutics for developmental and epileptic encephalopathies. Front Neurosci 2023; 17:1140679. [PMID: 37090807 PMCID: PMC10115946 DOI: 10.3389/fnins.2023.1140679] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/16/2023] [Indexed: 04/25/2023] Open
Abstract
Developmental and epileptic encephalopathies are childhood syndromes of severe epilepsy associated with cognitive and behavioral disorders. Of note, epileptic seizures represent only a part, although substantial, of the clinical spectrum. Whether the epileptiform activity per se accounts for developmental and intellectual disabilities is still unclear. In a few cases, seizures can be alleviated by antiseizure medication (ASM). However, the major comorbid features associated remain unsolved, including psychiatric disorders such as autism-like and attention deficit hyperactivity disorder-like behavior. Not surprisingly, the number of genes known to be involved is continuously growing, and genetically engineered rodent models are valuable tools for investigating the impact of gene mutations on local and distributed brain circuits. Despite the inconsistencies and problems arising in the generation and validation of the different preclinical models, those are unique and precious tools to identify new molecular targets, and essential to provide prospects for effective therapeutics.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Laboratory of Neuropsychopharmacology, Department of Neuroscience Rita Levi Montalcini, Institute of Neuroscience Cavalieri Ottolenghi (NICO), University of Turin, Torino, Italy
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute of Turin (NIT), Torino, Italy
- *Correspondence: Ilaria Bertocchi,
| | - Marco Cambiaghi
- Department Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Mazahir T. Hasan
- Laboratory of Brain Circuits Therapeutics, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque – Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
20
|
Schmitt LM, Li J, Liu R, Horn PS, Sweeney JA, Erickson CA, Pedapati EV. Altered frontal connectivity as a mechanism for executive function deficits in fragile X syndrome. Mol Autism 2022; 13:47. [PMID: 36494861 PMCID: PMC9733336 DOI: 10.1186/s13229-022-00527-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the leading inherited monogenic cause of intellectual disability and autism spectrum disorder. Executive function (EF), necessary for adaptive goal-oriented behavior and dependent on frontal lobe function, is impaired in individuals with FXS. Yet, little is known how alterations in frontal lobe neural activity is related to EF deficits in FXS. METHODS Sixty-one participants with FXS (54% males) and 71 age- and sex-matched typically-developing controls (TDC; 58% males) completed a five-minute resting state electroencephalography (EEG) protocol and a computerized battery of tests of EF, the Test of Attentional Performance for Children (KiTAP). Following source localization (minimum-norm estimate), we computed debiased weighted phase lag index (dWPLI), a phase connectivity value, for pairings between 18 nodes in frontal regions for gamma (30-55 Hz) and alpha (10.5-12.5 Hz) bands. Linear models were generated with fixed factors of group, sex, frequency, and connection. Relationships between frontal connectivity and EF variables also were examined. RESULTS Individuals with FXS demonstrated increased gamma band and reduced alpha band connectivity across all frontal regions and across hemispheres compared to TDC. After controlling for nonverbal IQ, increased error rates on EF tasks were associated with increased gamma band and reduced alpha band connectivity. LIMITATIONS Frontal connectivity findings are limited to intrinsic brain activity during rest and may not generalize to frontal connectivity during EF tasks or everyday function. CONCLUSIONS We report gamma hyper-connectivity and alpha hypo-connectivity within source-localized frontal brain regions in FXS compared to TDC during resting-state EEG. For the first time in FXS, we report significant associations between EF and altered frontal connectivity, with increased error rate relating to increased gamma band connectivity and reduced alpha band connectivity. These findings suggest increased phase connectivity within gamma band may impair EF performance, whereas greater alpha band connectivity may provide compensatory support for EF. Together, these findings provide important insight into neurophysiological mechanisms of EF deficits in FXS and provide novel targets for treatment development.
Collapse
Affiliation(s)
- Lauren M. Schmitt
- grid.239573.90000 0000 9025 8099Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 4002, Cincinnati, OH 45229 USA ,grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Joy Li
- grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Rui Liu
- grid.239573.90000 0000 9025 8099Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 4002, Cincinnati, OH 45229 USA
| | - Paul S. Horn
- grid.239573.90000 0000 9025 8099Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 4002, Cincinnati, OH 45229 USA ,grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - John A. Sweeney
- grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Craig A. Erickson
- grid.239573.90000 0000 9025 8099Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 4002, Cincinnati, OH 45229 USA ,grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Ernest V. Pedapati
- grid.239573.90000 0000 9025 8099Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 4002, Cincinnati, OH 45229 USA ,grid.24827.3b0000 0001 2179 9593University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
21
|
Kat R, Kas MJH. Largely unaffected auditory and visual sensory processing phenotypes in the evoked potentials of Fmr1 KO2 mice. Eur J Neurosci 2022; 56:5260-5273. [PMID: 36017614 PMCID: PMC9826194 DOI: 10.1111/ejn.15808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 01/11/2023]
Abstract
Sensory sensitivity symptoms are common in autism spectrum disorders and fragile X syndrome. Mainly in the auditory modality, disturbed processing has been found in both fragile X patients and the corresponding genetic mouse model, the Fmr1 knockout mouse. Here, we tried to replicate the auditory deficits and assess whether also visual processing is affected, using electroencephalography readouts under freely behaving conditions in the second-generation Fmr1 knockout mice. No differences between wild-type and knockout animals were found in single auditory and visual evoked potentials in response to pure sine tones and full-field light flashes. Visual sensory gating was enhanced in the early but not the late components of the evoked potentials, but no changes were found in auditory sensory gating. The higher harmonics of the synchronisation response to flickering visual stimuli seemed to be reduced with 10, but not 20 or 40 Hz, stimulation. However, this effect was not reproduced in an independent second cohort of animals. No synchronisation differences were found in response to a chirp stimulus, of which the frequency steadily increased. Taken together, this study could not reproduce earlier reported increased amplitudes in auditory responses, nor could it convincingly show that synchronisation deficits found to be present in the auditory modality also existed in the visual modality. The discrepancies within this study as well as between various studies assessing sensory processing in the Fmr1 KO raise questions about the external validity of these phenotypes and warrant careful interpretation of these phenotypes.
Collapse
Affiliation(s)
- Renate Kat
- Groningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| | - Martien J. H. Kas
- Groningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| |
Collapse
|
22
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
23
|
Wilde M, Constantin L, Thorne PR, Montgomery JM, Scott EK, Cheyne JE. Auditory processing in rodent models of autism: a systematic review. J Neurodev Disord 2022; 14:48. [PMID: 36042393 PMCID: PMC9429780 DOI: 10.1186/s11689-022-09458-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/07/2022] [Indexed: 11/19/2022] Open
Abstract
Autism is a complex condition with many traits, including differences in auditory sensitivity. Studies in human autism are plagued by the difficulty of controlling for aetiology, whereas studies in individual rodent models cannot represent the full spectrum of human autism. This systematic review compares results in auditory studies across a wide range of established rodent models of autism to mimic the wide range of aetiologies in the human population. A search was conducted in the PubMed and Web of Science databases to find primary research articles in mouse or rat models of autism which investigate central auditory processing. A total of 88 studies were included. These used non-invasive measures of auditory function, such as auditory brainstem response recordings, cortical event-related potentials, electroencephalography, and behavioural tests, which are translatable to human studies. They also included invasive measures, such as electrophysiology and histology, which shed insight on the origins of the phenotypes found in the non-invasive studies. The most consistent results across these studies were increased latency of the N1 peak of event-related potentials, decreased power and coherence of gamma activity in the auditory cortex, and increased auditory startle responses to high sound levels. Invasive studies indicated loss of subcortical inhibitory neurons, hyperactivity in the lateral superior olive and auditory thalamus, and reduced specificity of responses in the auditory cortex. This review compares the auditory phenotypes across rodent models and highlights those that mimic findings in human studies, providing a framework and avenues for future studies to inform understanding of the auditory system in autism.
Collapse
Affiliation(s)
- Maya Wilde
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter R Thorne
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Section of Audiology, School of Population Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.,Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Juliette E Cheyne
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
24
|
De Novo ZMYND8 variants result in an autosomal dominant neurodevelopmental disorder with cardiac malformations. Genet Med 2022; 24:1952-1966. [PMID: 35916866 DOI: 10.1016/j.gim.2022.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 12/25/2022] Open
Abstract
PURPOSE ZMYND8 encodes a multidomain protein that serves as a central interactive hub for coordinating critical roles in transcription regulation, chromatin remodeling, regulation of super-enhancers, DNA damage response and tumor suppression. We delineate a novel neurocognitive disorder caused by variants in the ZMYND8 gene. METHODS An international collaboration, exome sequencing, molecular modeling, yeast two-hybrid assays, analysis of available transcriptomic data and a knockdown Drosophila model were used to characterize the ZMYND8 variants. RESULTS ZMYND8 variants were identified in 11 unrelated individuals; 10 occurred de novo and one suspected de novo; 2 were truncating, 9 were missense, of which one was recurrent. The disorder is characterized by intellectual disability with variable cardiovascular, ophthalmologic and minor skeletal anomalies. Missense variants in the PWWP domain of ZMYND8 abolish the interaction with Drebrin and missense variants in the MYND domain disrupt the interaction with GATAD2A. ZMYND8 is broadly expressed across cell types in all brain regions and shows highest expression in the early stages of brain development. Neuronal knockdown of the DrosophilaZMYND8 ortholog results in decreased habituation learning, consistent with a role in cognitive function. CONCLUSION We present genomic and functional evidence for disruption of ZMYND8 as a novel etiology of syndromic intellectual disability.
Collapse
|
25
|
JA R, Lovelace JW, Kokash J, Hussain A, KA R. Nicotine reduces age-related changes in cortical neural oscillations without affecting auditory brainstem responses. Neurobiol Aging 2022; 120:10-26. [DOI: 10.1016/j.neurobiolaging.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 11/29/2022]
|
26
|
Bülow P, Segal M, Bassell GJ. Mechanisms Driving the Emergence of Neuronal Hyperexcitability in Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms23116315. [PMID: 35682993 PMCID: PMC9181819 DOI: 10.3390/ijms23116315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperexcitability is a shared neurophysiological phenotype across various genetic neurodevelopmental disorders, including Fragile X syndrome (FXS). Several patient symptoms are associated with hyperexcitability, but a puzzling feature is that their onset is often delayed until their second and third year of life. It remains unclear how and why hyperexcitability emerges in neurodevelopmental disorders. FXS is caused by the loss of FMRP, an RNA-binding protein which has many critical roles including protein synthesis-dependent and independent regulation of ion channels and receptors, as well as global regulation of protein synthesis. Here, we discussed recent literature uncovering novel mechanisms that may drive the progressive onset of hyperexcitability in the FXS brain. We discussed in detail how recent publications have highlighted defects in homeostatic plasticity, providing new insight on the FXS brain and suggest pharmacotherapeutic strategies in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| | - Menahem Segal
- Department of Brain Science, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Gary J. Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| |
Collapse
|
27
|
Targeted therapy of cognitive deficits in fragile X syndrome. Mol Psychiatry 2022; 27:2766-2776. [PMID: 35354925 PMCID: PMC7612812 DOI: 10.1038/s41380-022-01527-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/08/2022]
Abstract
Breaking an impasse in finding mechanism-based therapies of neuropsychiatric disorders requires a strategic shift towards alleviating individual symptoms. Here we present a symptom and circuit-specific approach to rescue deficits of reward learning in Fmr1 knockout mice, a model of Fragile X syndrome (FXS), the most common monogenetic cause of inherited mental disability and autism. We use high-throughput, ecologically-relevant automated tests of cognition and social behavior to assess effectiveness of the circuit-targeted injections of designer nanoparticles, loaded with TIMP metalloproteinase inhibitor 1 protein (TIMP-1). Further, to investigate the impact of our therapeutic strategy on neuronal plasticity we perform long-term potentiation recordings and high-resolution electron microscopy. We show that central amygdala-targeted delivery of TIMP-1 designer nanoparticles reverses impaired cognition in Fmr1 knockouts, while having no impact on deficits of social behavior, hence corroborating symptom-specificity of the proposed approach. Moreover, we elucidate the neural correlates of the highly specific behavioral rescue by showing that the applied therapeutic intervention restores functional synaptic plasticity and ultrastructure of neurons in the central amygdala. Thus, we present a targeted, symptom-specific and mechanism-based strategy to remedy cognitive deficits in Fragile X syndrome.
Collapse
|
28
|
Holley A, Shedd A, Boggs A, Lovelace J, Erickson C, Gross C, Jankovic M, Razak K, Huber K, Gibson JR. A sound-driven cortical phase-locking change in the Fmr1 KO mouse requires Fmr1 deletion in a subpopulation of brainstem neurons. Neurobiol Dis 2022; 170:105767. [PMID: 35588990 PMCID: PMC9273231 DOI: 10.1016/j.nbd.2022.105767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/27/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Sensory impairments commonly occur in patients with autism or intellectual disability. Fragile X syndrome (FXS) is one form of intellectual disability that is often comorbid with autism. In electroencephalographic (EEG) recordings obtained from humans with FXS, the ability of cortical regions to consistently synchronize, or “phase-lock”, to modulated auditory stimuli is reduced compared to that of typically developing individuals. At the same time, less time-locked, “non-phase-locked” power induced by sounds is higher. The same changes occur in the Fmr1 knockout (KO) mouse – an animal model of FXS. We determined if Fmr1 deletion in a subset of brainstem auditory neurons plays any role in these EEG changes in the mouse. Methods: We reinstated FMRP expression in a subpopulation of brainstem auditory neurons in an otherwise Fmr1 KO control (conditional on; cON Fmr1) mouse and used EEG recordings to determine if reinstatement normalized, or “rescued”, the phase-locking phenotype observed in the cON Fmr1 mouse. In determining rescue, this also meant that Fmr1 deletion in the same neuron population was necessary for the phenotype to occur. Results: We find that Fmr1 reinstatement in a subset of brainstem neurons rescues certain aspects of the phase-locking phenotype but does not rescue the increase in non-phase-locked power. Unexpectedly, not all electrophysiological phenotypes observed in the Fmr1 KO were observed in the cON Fmr1 mouse used for the reinstatement experiments, and this was likely due to residual expression of FMRP in these Fmr1 KO controls. Conclusions: Fmr1 deletion in brainstem neurons is necessary for certain aspects of the decreased phase-locking phenotype in the Fmr1 KO, but not necessary for the increase in non-phase-locked power induced by a sound. The most likely brainstem structure underlying these results is the inferior colliculus. We also demonstrate that low levels of FMRP can rescue some EEG phenotypes but not others. This latter finding provides a foundation for how symptoms in FXS individuals may vary due to FMRP levels and that reinstatement of low FMRP levels may be sufficient to alleviate particular symptoms.
Collapse
Affiliation(s)
- AndrewJ Holley
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Aleya Shedd
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Anna Boggs
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jonathan Lovelace
- Department of Psychology, University of California, Riverside, CA 92521, USA
| | - Craig Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Miranda Jankovic
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Khaleel Razak
- Department of Psychology, University of California, Riverside, CA 92521, USA
| | - Kimberly Huber
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Jay R Gibson
- University of Texas Southwestern Medical Center at Dallas, Department of Neuroscience, Dallas, TX 75390-9111, USA.
| |
Collapse
|
29
|
Scaramella C, Alzagatiti JB, Creighton C, Mankatala S, Licea F, Winter GM, Emtage J, Wisnieski JR, Salazar L, Hussain A, Lee FM, Mammootty A, Mammootty N, Aldujaili A, Runnberg KA, Hernandez D, Zimmerman-Thompson T, Makwana R, Rouvere J, Tahmasebi Z, Zavradyan G, Campbell CS, Komaranchath M, Carmona J, Trevitt J, Glanzman D, Roberts AC. Bisphenol A Exposure Induces Sensory Processing Deficits in Larval Zebrafish during Neurodevelopment. eNeuro 2022; 9:ENEURO.0020-22.2022. [PMID: 35508370 PMCID: PMC9116930 DOI: 10.1523/eneuro.0020-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/10/2022] [Accepted: 04/01/2022] [Indexed: 11/21/2022] Open
Abstract
Because of their ex utero development, relatively simple nervous system, translucency, and availability of tools to investigate neural function, larval zebrafish are an exceptional model for understanding neurodevelopmental disorders and the consequences of environmental toxins. Furthermore, early in development, zebrafish larvae easily absorb chemicals from water, a significant advantage over methods required to expose developing organisms to chemical agents in utero Bisphenol A (BPA) and BPA analogs are ubiquitous environmental toxins with known molecular consequences. All humans have measurable quantities of BPA in their bodies. Most concerning, the level of BPA exposure is correlated with neurodevelopmental difficulties in people. Given the importance of understanding the health-related effects of this common toxin, we have exploited the experimental advantages of the larval zebrafish model system to investigate the behavioral and anatomic effects of BPA exposure. We discovered that BPA exposure early in development leads to deficits in the processing of sensory information, as indicated by BPA's effects on prepulse inhibition (PPI) and short-term habituation (STH) of the C-start reflex. We observed no changes in locomotion, thigmotaxis, and repetitive behaviors (circling). Despite changes in sensory processing, we detected no regional or whole-brain volume changes. Our results show that early BPA exposure can induce sensory processing deficits, as revealed by alterations in simple behaviors that are mediated by a well-defined neural circuit.
Collapse
Affiliation(s)
- Courtney Scaramella
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Joseph B Alzagatiti
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Christopher Creighton
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Samandeep Mankatala
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Fernando Licea
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Gabriel M Winter
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Jasmine Emtage
- Department of Biology, California Institute of Technology, Pasadena, CA 91125
| | - Joseph R Wisnieski
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Luis Salazar
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Anjum Hussain
- Department of Neuroscience, University of California, Riverside, Riverside, CA 92521
| | - Faith M Lee
- Department of Society and Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Asma Mammootty
- Saint Louis University School of Medicine, St. Louis, MO 63104
| | | | - Andrew Aldujaili
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Kristine A Runnberg
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Daniela Hernandez
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | | | - Rikhil Makwana
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Julien Rouvere
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Zahra Tahmasebi
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Gohar Zavradyan
- Department of Neuroscience, University of California, Riverside, Riverside, CA 92521
| | | | - Meghna Komaranchath
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Javier Carmona
- Department of Physics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Jennifer Trevitt
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - David Glanzman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
- Integrative Center for Learning and Memory, Brain Research Institute, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Adam C Roberts
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| |
Collapse
|
30
|
Castro AC, Monteiro P. Auditory Dysfunction in Animal Models of Autism Spectrum Disorder. Front Mol Neurosci 2022; 15:845155. [PMID: 35493332 PMCID: PMC9043325 DOI: 10.3389/fnmol.2022.845155] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder mainly characterized by social-communication impairments, repetitive behaviors and altered sensory perception. Auditory hypersensitivity is the most common sensory-perceptual abnormality in ASD, however, its underlying neurobiological mechanisms remain elusive. Consistently with reports in ASD patients, animal models for ASD present sensory-perception alterations, including auditory processing impairments. Here we review the current knowledge regarding auditory dysfunction in rodent models of ASD, exploring both shared and distinct features among them, mechanistic and molecular underpinnings, and potential therapeutic approaches. Overall, auditory dysfunction in ASD models seems to arise from impaired central processing. Depending on the model, impairments may arise at different steps along the auditory pathway, from auditory brainstem up to the auditory cortex. Common defects found across models encompass atypical tonotopicity in different regions of the auditory pathway, temporal and spectral processing impairments and histological differences. Imbalance between excitation and inhibition (E/I imbalance) is one of the most well-supported mechanisms explaining the auditory phenotype in the ASD models studied so far and seems to be linked to alterations in GABAergic signaling. Such E/I imbalance may have a large impact on the development of the auditory pathway, influencing the establishment of connections responsible for normal sound processing.
Collapse
Affiliation(s)
- Ana Carolina Castro
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, Portugal
| | - Patricia Monteiro
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
31
|
Marquez-Legorreta E, Constantin L, Piber M, Favre-Bulle IA, Taylor MA, Blevins AS, Giacomotto J, Bassett DS, Vanwalleghem GC, Scott EK. Brain-wide visual habituation networks in wild type and fmr1 zebrafish. Nat Commun 2022; 13:895. [PMID: 35173170 PMCID: PMC8850451 DOI: 10.1038/s41467-022-28299-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Habituation is a form of learning during which animals stop responding to repetitive stimuli, and deficits in habituation are characteristic of several psychiatric disorders. Due to technical challenges, the brain-wide networks mediating habituation are poorly understood. Here we report brain-wide calcium imaging during larval zebrafish habituation to repeated visual looming stimuli. We show that different functional categories of loom-sensitive neurons are located in characteristic locations throughout the brain, and that both the functional properties of their networks and the resulting behavior can be modulated by stimulus saliency and timing. Using graph theory, we identify a visual circuit that habituates minimally, a moderately habituating midbrain population proposed to mediate the sensorimotor transformation, and downstream circuit elements responsible for higher order representations and the delivery of behavior. Zebrafish larvae carrying a mutation in the fmr1 gene have a systematic shift toward sustained premotor activity in this network, and show slower behavioral habituation.
Collapse
Affiliation(s)
- Emmanuel Marquez-Legorreta
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Marielle Piber
- School of Medicine, Medical Sciences, and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Itia A Favre-Bulle
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,School of Mathematics and Physics, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Michael A Taylor
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ann S Blevins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Giacomotto
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD, 4076, Australia.,Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.,Discovery Biology, Griffith University, Brisbane, QLD, 4111, Australia
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Departments of Electrical & Systems Engineering, Physics & Astronomy, Neurology, Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
32
|
Kenny A, Wright D, Stanfield AC. EEG as a translational biomarker and outcome measure in fragile X syndrome. Transl Psychiatry 2022; 12:34. [PMID: 35075104 PMCID: PMC8786970 DOI: 10.1038/s41398-022-01796-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/01/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
Targeted treatments for fragile X syndrome (FXS) have frequently failed to show efficacy in clinical testing, despite success at the preclinical stages. This has highlighted the need for more effective translational outcome measures. EEG differences observed in FXS, including exaggerated N1 ERP amplitudes, increased resting gamma power and reduced gamma phase-locking in the sensory cortices, have been suggested as potential biomarkers of the syndrome. These abnormalities are thought to reflect cortical hyper excitability resulting from an excitatory (glutamate) and inhibitory (GABAergic) imbalance in FXS, which has been the target of several pharmaceutical remediation studies. EEG differences observed in humans also show similarities to those seen in laboratory models of FXS, which may allow for greater translational equivalence and better predict clinical success of putative therapeutics. There is some evidence from clinical trials showing that treatment related changes in EEG may be associated with clinical improvements, but these require replication and extension to other medications. Although the use of EEG characteristics as biomarkers is still in the early phases, and further research is needed to establish its utility in clinical trials, the current research is promising and signals the emergence of an effective translational biomarker.
Collapse
Affiliation(s)
- Aisling Kenny
- Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF, Edinburgh, UK.
| | - Damien Wright
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| | - Andrew C. Stanfield
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| |
Collapse
|
33
|
Liu X, Kumar V, Tsai NP, Auerbach BD. Hyperexcitability and Homeostasis in Fragile X Syndrome. Front Mol Neurosci 2022; 14:805929. [PMID: 35069112 PMCID: PMC8770333 DOI: 10.3389/fnmol.2021.805929] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 01/13/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading inherited cause of autism and intellectual disability, resulting from a mutation in the FMR1 gene and subsequent loss of its protein product FMRP. Despite this simple genetic origin, FXS is a phenotypically complex disorder with a range of physical and neurocognitive disruptions. While numerous molecular and cellular pathways are affected by FMRP loss, there is growing evidence that circuit hyperexcitability may be a common convergence point that can account for many of the wide-ranging phenotypes seen in FXS. The mechanisms for hyperexcitability in FXS include alterations to excitatory synaptic function and connectivity, reduced inhibitory neuron activity, as well as changes to ion channel expression and conductance. However, understanding the impact of FMR1 mutation on circuit function is complicated by the inherent plasticity in neural circuits, which display an array of homeostatic mechanisms to maintain activity near set levels. FMRP is also an important regulator of activity-dependent plasticity in the brain, meaning that dysregulated plasticity can be both a cause and consequence of hyperexcitable networks in FXS. This makes it difficult to separate the direct effects of FMR1 mutation from the myriad and pleiotropic compensatory changes associated with it, both of which are likely to contribute to FXS pathophysiology. Here we will: (1) review evidence for hyperexcitability and homeostatic plasticity phenotypes in FXS models, focusing on similarities/differences across brain regions, cell-types, and developmental time points; (2) examine how excitability and plasticity disruptions interact with each other to ultimately contribute to circuit dysfunction in FXS; and (3) discuss how these synaptic and circuit deficits contribute to disease-relevant behavioral phenotypes like epilepsy and sensory hypersensitivity. Through this discussion of where the current field stands, we aim to introduce perspectives moving forward in FXS research.
Collapse
Affiliation(s)
- Xiaopeng Liu
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Vipendra Kumar
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Nien-Pei Tsai
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Benjamin D. Auerbach
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Benjamin D. Auerbach
| |
Collapse
|
34
|
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, Ethell IM. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome. Neurobiol Dis 2022; 162:105577. [PMID: 34871737 DOI: 10.1016/j.nbd.2021.105577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement. METHODS To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated CreCaMKIIa/Fmr1Flox/y (cOFF) and CreCaMKIIa/Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests. RESULTS We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved. CONCLUSIONS These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS.
Collapse
Affiliation(s)
- Maham Rais
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Xinghao S Shuai
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Walker Woodard
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Steven Bishay
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Leo Estrada
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Ashwin R Sharma
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Austin Nguy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Patricia S Pirbhoy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Arnold R Palacios
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | | | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
35
|
Contractor A, Ethell IM, Portera-Cailliau C. Cortical interneurons in autism. Nat Neurosci 2021; 24:1648-1659. [PMID: 34848882 PMCID: PMC9798607 DOI: 10.1038/s41593-021-00967-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
The mechanistic underpinnings of autism remain a subject of debate and controversy. Why do individuals with autism share an overlapping set of atypical behaviors and symptoms, despite having different genetic and environmental risk factors? A major challenge in developing new therapies for autism has been the inability to identify convergent neural phenotypes that could explain the common set of symptoms that result in the diagnosis. Although no striking macroscopic neuropathological changes have been identified in autism, there is growing evidence that inhibitory interneurons (INs) play an important role in its neural basis. In this Review, we evaluate and interpret this evidence, focusing on recent findings showing reduced density and activity of the parvalbumin class of INs. We discuss the need for additional studies that investigate how genes and the environment interact to change the developmental trajectory of INs, permanently altering their numbers, connectivity and circuit engagement.
Collapse
Affiliation(s)
- Anis Contractor
- Department of Neuroscience Feinberg School of Medicine, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, UC Riverside School of Medicine, Riverside, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Rumschlag JA, Razak KA. Age-related changes in event related potentials, steady state responses and temporal processing in the auditory cortex of mice with severe or mild hearing loss. Hear Res 2021; 412:108380. [PMID: 34758398 DOI: 10.1016/j.heares.2021.108380] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 08/19/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022]
Abstract
Age-related changes in auditory processing affect the quality of life of older adults with and without hearing loss. To distinguish between the effects of sensorineural hearing loss and aging on cortical processing, the main goal of the present study was to compare cortical responses using the same stimulus paradigms and recording conditions in two strains of mice (C57BL/6J and FVB) that differ in the degree of age-related hearing loss. Electroencephalogram (EEG) recordings were obtained from freely moving young and old mice using epidural screw electrodes. We measured event related potentials (ERP) and 40 Hz auditory steady-state responses (ASSR). We used a novel stimulus, termed the gap-ASSR stimulus, which elicits an ASSR by rapidly presenting short gaps in continuous noise. By varying the gap widths and modulation depths, we probed the limits of temporal processing in young and old mice. Temporal fidelity of ASSR and gap-ASSR responses were measured as phase consistency across trials (inter-trial phase clustering; ITPC). The old C57 mice, which show severe hearing loss, produced larger ERP amplitudes compared to young mice. Despite robust ERPs, the old C57 mice showed significantly diminished ITPC in the ASSR and gap-ASSR responses, even with 100% modulation depth. The FVB mice, which show mild hearing loss with age, generated similar ERP amplitudes and ASSR ITPC across the age groups tested. However, the old FVB mice showed decreased gap-ASSR responses compared to young mice, particularly for modulation depths <100%. The C57 mice data suggest that severe presbycusis leads to increased gain in the auditory cortex, but with reduced temporal fidelity. The FVB mice data suggest that with mild hearing loss, age-related changes in temporal processing become apparent only when tested with more challenging sounds (shorter gaps and shallower modulation).
Collapse
Affiliation(s)
| | - Khaleel A Razak
- Graduate Neuroscience Program, Riverside, United States; Psychology Department, University of California, Riverside, United States.
| |
Collapse
|
37
|
Razak KA, Binder DK, Ethell IM. Neural Correlates of Auditory Hypersensitivity in Fragile X Syndrome. Front Psychiatry 2021; 12:720752. [PMID: 34690832 PMCID: PMC8529206 DOI: 10.3389/fpsyt.2021.720752] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/16/2021] [Indexed: 01/20/2023] Open
Abstract
The mechanisms underlying the common association between autism spectrum disorders (ASD) and sensory processing disorders (SPD) are unclear, and treatment options to reduce atypical sensory processing are limited. Fragile X Syndrome (FXS) is a leading genetic cause of intellectual disability and ASD behaviors. As in most children with ASD, atypical sensory processing is a common symptom in FXS, frequently manifesting as sensory hypersensitivity. Auditory hypersensitivity is a highly debilitating condition in FXS that may lead to language delays, social anxiety and ritualized repetitive behaviors. Animal models of FXS, including Fmr1 knock out (KO) mouse, also show auditory hypersensitivity, providing a translation relevant platform to study underlying pathophysiological mechanisms. The focus of this review is to summarize recent studies in the Fmr1 KO mouse that identified neural correlates of auditory hypersensitivity. We review results of electroencephalography (EEG) recordings in the Fmr1 KO mice and highlight EEG phenotypes that are remarkably similar to EEG findings in humans with FXS. The EEG phenotypes associated with the loss of FMRP include enhanced resting EEG gamma band power, reduced cross frequency coupling, reduced sound-evoked synchrony of neural responses at gamma band frequencies, increased event-related potential amplitudes, reduced habituation of neural responses and increased non-phase locked power. In addition, we highlight the postnatal period when the EEG phenotypes develop and show a strong association of the phenotypes with enhanced matrix-metalloproteinase-9 (MMP-9) activity, abnormal development of parvalbumin (PV)-expressing inhibitory interneurons and reduced formation of specialized extracellular matrix structures called perineuronal nets (PNNs). Finally, we discuss how dysfunctions of inhibitory PV interneurons may contribute to cortical hyperexcitability and EEG abnormalities observed in FXS. Taken together, the studies reviewed here indicate that EEG recordings can be utilized in both pre-clinical studies and clinical trials, while at the same time, used to identify cellular and circuit mechanisms of dysfunction in FXS. New therapeutic approaches that reduce MMP-9 activity and restore functions of PV interneurons may succeed in reducing FXS sensory symptoms. Future studies should examine long-lasting benefits of developmental vs. adult interventions on sensory phenotypes.
Collapse
Affiliation(s)
- Khaleel A. Razak
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K. Binder
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M. Ethell
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
38
|
Liu DC, Lee KY, Lizarazo S, Cook JK, Tsai NP. ER stress-induced modulation of neural activity and seizure susceptibility is impaired in a fragile X syndrome mouse model. Neurobiol Dis 2021; 158:105450. [PMID: 34303799 DOI: 10.1016/j.nbd.2021.105450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/18/2021] [Indexed: 01/29/2023] Open
Abstract
Imbalanced neuronal excitability homeostasis is commonly observed in patients with fragile X syndrome (FXS) and the animal model of FXS, the Fmr1 KO. While alterations of neuronal intrinsic excitability and synaptic activity at the steady state in FXS have been suggested to contribute to such a deficit and ultimately the increased susceptibility to seizures in FXS, it remains largely unclear whether and how the homeostatic response of neuronal excitability following extrinsic challenges is disrupted in FXS. Our previous work has shown that the acute response following induction of endoplasmic reticulum (ER) stress can reduce neural activity and seizure susceptibility. Because many signaling pathways associated with ER stress response are mediated by Fmr1, we asked whether acute ER stress-induced reduction of neural activity and seizure susceptibility are altered in FXS. Our results first revealed that acute ER stress can trigger a protein synthesis-dependent prevention of neural network synchronization in vitro and a reduction of susceptibility to kainic acid-induced seizures in vivo in wild-type but not in Fmr1 KO mice. Mechanistically, we found that acute ER stress-induced activation of murine double minute-2 (Mdm2), ubiquitination of p53, and the subsequent transient protein synthesis are all impaired in Fmr1 KO neurons. Employing a p53 inhibitor, Pifithrin-α, to mimic p53 inactivation, we were able to blunt the increase in neural network synchronization and reduce the seizure susceptibility in Fmr1 KO mice following ER stress induction. In summary, our data revealed a novel cellular defect in Fmr1 KO mice and suggest that an impaired response to common extrinsic challenges may contribute to imbalanced neuronal excitability homeostasis in FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jessie K Cook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nien-Pei Tsai
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
39
|
Lovelace JW, Rais M, Palacios AR, Shuai XS, Bishay S, Popa O, Pirbhoy PS, Binder DK, Nelson DL, Ethell IM, Razak KA. Deletion of Fmr1 from Forebrain Excitatory Neurons Triggers Abnormal Cellular, EEG, and Behavioral Phenotypes in the Auditory Cortex of a Mouse Model of Fragile X Syndrome. Cereb Cortex 2021; 30:969-988. [PMID: 31364704 DOI: 10.1093/cercor/bhz141] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Fragile X syndrome (FXS) is a leading genetic cause of autism with symptoms that include sensory processing deficits. In both humans with FXS and a mouse model [Fmr1 knockout (KO) mouse], electroencephalographic (EEG) recordings show enhanced resting state gamma power and reduced sound-evoked gamma synchrony. We previously showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to these phenotypes by affecting perineuronal nets (PNNs) around parvalbumin (PV) interneurons in the auditory cortex of Fmr1 KO mice. However, how different cell types within local cortical circuits contribute to these deficits is not known. Here, we examined whether Fmr1 deletion in forebrain excitatory neurons affects neural oscillations, MMP-9 activity, and PV/PNN expression in the auditory cortex. We found that cortical MMP-9 gelatinase activity, mTOR/Akt phosphorylation, and resting EEG gamma power were enhanced in CreNex1/Fmr1Flox/y conditional KO (cKO) mice, whereas the density of PV/PNN cells was reduced. The CreNex1/Fmr1Flox/y cKO mice also show increased locomotor activity, but not the anxiety-like behaviors. These results indicate that fragile X mental retardation protein changes in excitatory neurons in the cortex are sufficient to elicit cellular, electrophysiological, and behavioral phenotypes in Fmr1 KO mice. More broadly, these results indicate that local cortical circuit abnormalities contribute to sensory processing deficits in autism spectrum disorders.
Collapse
Affiliation(s)
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine
| | | | | | | | - Otilia Popa
- Division of Biomedical Sciences, School of Medicine
| | | | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - David L Nelson
- Molecular and Human Genetics, Baylor College of Medicine , Houston, TX 77030, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - Khaleel A Razak
- Department of Psychology.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| |
Collapse
|
40
|
Lee CH, Kim KW, Lee DH, Lee SM, Kim SY. Overexpression of the receptor for advanced glycation end-products in the auditory cortex of rats with noise-induced hearing loss. BMC Neurosci 2021; 22:38. [PMID: 34020590 PMCID: PMC8139161 DOI: 10.1186/s12868-021-00642-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The receptor for advanced glycation end-products (RAGE) is involved in neuroinflammation. This study investigated the changes in RAGE expression following noise-induced hearing loss. METHODS Three-week-old female Sprague-Dawley rats were exposed to 115 dB SPL white noise for 4 h daily for 3 d (noise group, n = 16). In parallel, age and sex-matched control rats were raised under standard conditions without noise exposure (control group, n = 16). After 2 h (noise immediate, n = 8) and 4 wk (noise 4-week, n = 8) of noise exposure, the auditory cortex was harvested and cytoplasmic and nuclear fractions were isolated. The gene expression levels of tumor necrosis factor alpha (TNF-α), interleukin 6 (IL6), interleukin 1 beta (IL1β), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and RAGE were evaluated using real-time reverse transcription polymerase chain reaction. The protein expression levels of nuclear RAGE and cytosolic RAGE were evaluated using western blotting. Additionally, matrix metalloproteinase 9 (MMP9) was pharmacologically inhibited in the noise immediate group, and then nuclear and cytosolic RAGE expression levels were evaluated. RESULTS The noise immediate and noise 4-week groups exhibited increased auditory thresholds at 4, 8, 16, and 32 kHz frequencies. The genes encoding the pro-inflammatory cytokines TNF-α, IL6, IL1β, and NF- κB were increased 3.74, 1.63, 6.42, and 6.23-fold in the noise immediate group, respectively (P = 0.047, 0.043, 0.044, and 0.041). RAGE mRNA expression was elevated 1.42-fold in the noise 4-week group (P = 0.032). Cytosolic RAGE expression was increased 1.76 and 6.99-fold in the noise immediate and noise 4-week groups, respectively (P = 0.04 and 0.03). Nuclear RAGE expression was comparable between the noise and control groups. matrix metalloproteinase 9 (MMP9) inhibition reduced cytosolic RAGE expression in the noise immediate group (P = 0.004). CONCLUSIONS Noise exposure increased the expression of cytosolic RAGE in the auditory cortex and upregulated pro-inflammatory genes, but this response could be alleviated by MMP9 inhibition.
Collapse
Affiliation(s)
- Chang Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University College of Medicine, 59, Yatap-ro, Bundang-gu, Seongnam, 13496, Gyeonggi-do, Korea
| | - Kyung Woon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University College of Medicine, 59, Yatap-ro, Bundang-gu, Seongnam, 13496, Gyeonggi-do, Korea
| | - Da-Hye Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University College of Medicine, 59, Yatap-ro, Bundang-gu, Seongnam, 13496, Gyeonggi-do, Korea
| | - So Min Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University College of Medicine, 59, Yatap-ro, Bundang-gu, Seongnam, 13496, Gyeonggi-do, Korea
| | - So Young Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University College of Medicine, 59, Yatap-ro, Bundang-gu, Seongnam, 13496, Gyeonggi-do, Korea.
| |
Collapse
|
41
|
Mascio G, Bucci D, Notartomaso S, Liberatore F, Antenucci N, Scarselli P, Imbriglio T, Caruso S, Gradini R, Cannella M, Di Menna L, Bruno V, Battaglia G, Nicoletti F. Perineuronal nets are under the control of type-5 metabotropic glutamate receptors in the developing somatosensory cortex. Transl Psychiatry 2021; 11:109. [PMID: 33597513 PMCID: PMC7889908 DOI: 10.1038/s41398-021-01210-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
mGlu5 metabotropic glutamate receptors are highly functional in the early postnatal life, and regulate developmental plasticity of parvalbumin-positive (PV+) interneurons in the cerebral cortex. PV+ cells are enwrapped by perineuronal nets (PNNs) at the closure of critical windows of cortical plasticity. Changes in PNNs have been associated with neurodevelopmental disorders. We found that the number of Wisteria Fluoribunda Agglutinin (WFA)+ PNNs and the density of WFA+/PV+ cells were largely increased in the somatosensory cortex of mGlu5-/- mice at PND16. An increased WFA+ PNN density was also observed after pharmacological blockade of mGlu5 receptors in the first two postnatal weeks. The number of WFA+ PNNs in mGlu5-/- mice was close to a plateau at PND16, whereas continued to increase in wild-type mice, and there was no difference between the two genotypes at PND21 and PND60. mGlu5-/- mice at PND16 showed increases in the transcripts of genes involved in PNN formation and a reduced expression and activity of type-9 matrix metalloproteinase in the somatosensory cortex suggesting that mGlu5 receptors control both PNN formation and degradation. Finally, unilateral whisker stimulation from PND9 to PND16 enhanced WFA+ PNN density in the contralateral somatosensory cortex only in mGlu5+/+ mice, whereas whisker trimming from PND9 to PND16 reduced WFA+ PNN density exclusively in mGlu5-/- mice, suggesting that mGlu5 receptors shape the PNN response to sensory experience. These findings disclose a novel undescribed mechanism of PNN regulation, and lay the groundwork for the study of mGlu5 receptors and PNNs in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Giada Mascio
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Domenico Bucci
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | | | | | - Nico Antenucci
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | | | - Stefano Caruso
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Roberto Gradini
- grid.7841.aDepartment of Experimental Medicine, Sapienza University, Rome, Italy
| | - Milena Cannella
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Luisa Di Menna
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Valeria Bruno
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy. .,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
42
|
Ethridge L, Thaliath A, Kraff J, Nijhawan K, Berry-Kravis E. Development of Neural Response to Novel Sounds in Fragile X Syndrome: Potential Biomarkers. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2020; 125:449-464. [PMID: 33211818 PMCID: PMC8631234 DOI: 10.1352/1944-7558-125.6.449] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Auditory processing abnormalities in fragile X syndrome (FXS) may contribute to difficulties with language development, pattern identification, and contextual updating. Participants with FXS (N = 41) and controls (N = 27) underwent auditory event-related potentials during presentation of an oddball paradigm. Data was adequate for analysis for 33 participants with FXS and 27 controls (age 4-51 y, 13 females [FXS]; 4-54 y, 11 females [control]). Participants with FXS showed larger N1 and P2 amplitudes, abnormal lack of modulation of P1 and P2 amplitudes and P2 latency in response to oddball stimuli ) relative to controls: Females with FXS were more similar to controls. Participants with FXS showed a marginal speeding of the P2 latency, suggesting potentiation to oddball stimuli rather than habituation. Participants with FXS showed a heightened N1 habituation effect compared to controls. Gamma power was significantly higher for participants with FXS. Groups did not differ on mismatch negativity. Both controls and participants with FXS showed similar developmental trajectories in P1 and N1 amplitude, P2 latency, and gamma power, but not for P2 amplitude. One month retest analyses performed in 14 participants suggest strong test-retest reliability for most measures. Individuals with FXS show previously demonstrated increased response amplitude and high frequency neural activity. Despite an overall normal developmental trajectory for most measures, individuals with FXS show age-independent but gender-dependent decreases in complex processing of novel stimuli. Many markers show strong retest reliability even in children and thus are potential biomarkers for clinical trials in FXS.
Collapse
Affiliation(s)
- Lauren Ethridge
- Lauren Ethridge, University of Oklahoma Health Sciences Center
| | - Andrew Thaliath
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Jeremy Kraff
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Karan Nijhawan
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Elizabeth Berry-Kravis
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| |
Collapse
|
43
|
FMR1 loss in a human stem cell model reveals early changes to intrinsic membrane excitability. Dev Biol 2020; 468:93-100. [PMID: 32976839 DOI: 10.1016/j.ydbio.2020.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 11/21/2022]
Abstract
Fragile X mental retardation 1 (FMR1) encodes the RNA binding protein FMRP. Loss of FMRP drives Fragile X syndrome (FXS), the leading inherited cause of intellectual disability and a leading monogenic cause of autism. While cortical hyperexcitability is a hallmark of FXS, the reported phenotypes and underlying mechanisms, including alterations in synaptic transmission and ion channel properties, are heterogeneous and at times contradictory. Here, we report the generation of new isogenic FMR1y/+ and FMR1y/- human pluripotent stem cell (hPSC) lines using CRISPR-Cas9 to facilitate the study of how complete FMRP loss, independent of genetic background, drives molecular and cellular alterations relevant for FXS. After differentiating these stem cell tools into excitatory neurons, we systematically assessed the impact of FMRP loss on intrinsic membrane and synaptic properties over time. Using whole-cell patch clamp analyses, we found that FMR1y/- neurons overall showed an increased intrinsic membrane excitability compared to age-matched FMR1y/+ controls, with no discernable alternations in synaptic transmission. Surprisingly, longitudinal analyses of cell intrinsic defects revealed that a majority of significant changes emerged early following in vitro differentiation and some were not stable over time. Collectively, this study provides a new isogenic hPSC model which can be further leveraged by the scientific community to investigate basic mechanisms of FMR1 gene function relevant for FXS. Moreover, our results suggest that precocious changes in the intrinsic membrane properties during early developmental could be a critical cellular pathology ultimately contributing to cortical hyperexcitability in FXS.
Collapse
|
44
|
Dong HW, Erickson K, Lee JR, Merritt J, Fu C, Neul JL. Detection of neurophysiological features in female R255X MeCP2 mutation mice. Neurobiol Dis 2020; 145:105083. [PMID: 32927061 PMCID: PMC7572861 DOI: 10.1016/j.nbd.2020.105083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/11/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder (NDD) that is nearly always caused by loss of function mutations in Methyl-CpG-binding Protein 2 (MECP2) and shares many clinical features with other NDD. Genetic restoration of Mecp2 in symptomatic mice lacking MeCP2 expression can reverse symptoms, providing hope that disease modifying therapies can be identified for RTT. Effective and rapid clinical trial completion relies on well-defined clinical outcome measures and robust biomarkers of treatment responses. Studies on other NDD have found evidence of differences in neurophysiological measures that correlate with disease severity. However, currently there are no well-validated biomarkers in RTT to predict disease prognosis or treatment responses. To address this, we characterized neurophysiological features in a mouse model of RTT containing a knock-in nonsense mutation (p.R255X) in the Mecp2 locus. We found a variety of changes in heterozygous female Mecp2R255X/X mice including age-related changes in sleep/wake architecture, alterations in baseline EEG power, increased incidence of spontaneous epileptiform discharges, and changes in auditory evoked potentials. Furthermore, we identified association of some neurophysiological features with disease severity. These findings provide a set of potential non-invasive and translatable biomarkers that can be utilized in preclinical therapy trials in animal models of RTT and eventually within the context of clinical trials.
Collapse
Affiliation(s)
- Hong-Wei Dong
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| | - Kirsty Erickson
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| | - Jessica R Lee
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| | - Jonathan Merritt
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| | - Cary Fu
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| | - Jeffrey L Neul
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA.
| |
Collapse
|
45
|
Lovelace JW, Ethell IM, Binder DK, Razak KA. Minocycline Treatment Reverses Sound Evoked EEG Abnormalities in a Mouse Model of Fragile X Syndrome. Front Neurosci 2020; 14:771. [PMID: 32848552 PMCID: PMC7417521 DOI: 10.3389/fnins.2020.00771] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability. Many symptoms of FXS overlap with those in autism including repetitive behaviors, language delays, anxiety, social impairments and sensory processing deficits. Electroencephalogram (EEG) recordings from humans with FXS and an animal model, the Fmr1 knockout (KO) mouse, show remarkably similar phenotypes suggesting that EEG phenotypes can serve as biomarkers for developing treatments. This includes enhanced resting gamma band power and sound evoked total power, and reduced fidelity of temporal processing and habituation of responses to repeated sounds. Given the therapeutic potential of the antibiotic minocycline in humans with FXS and animal models, it is important to determine sensitivity and selectivity of EEG responses to minocycline. Therefore, in this study, we examined if a 10-day treatment of adult Fmr1 KO mice with minocycline (oral gavage, 30 mg/kg per day) would reduce EEG abnormalities. We tested if minocycline treatment has specific effects based on the EEG measurement type (e.g., resting versus sound-evoked) from the frontal and auditory cortex of the Fmr1 KO mice. We show increased resting EEG gamma power and reduced phase locking to time varying stimuli as well as the 40 Hz auditory steady state response in the Fmr1 KO mice in the pre-drug condition. Minocycline treatment increased gamma band phase locking in response to auditory stimuli, and reduced sound-evoked power of auditory event related potentials (ERP) in Fmr1 KO mice compared to vehicle treatment. Minocycline reduced resting EEG gamma power in Fmr1 KO mice, but this effect was similar to vehicle treatment. We also report frequency band-specific effects on EEG responses. Taken together, these data indicate that sound-evoked EEG responses may serve as more sensitive measures, compared to resting EEG measures, to isolate minocycline effects from placebo in humans with FXS. Given the use of minocycline and EEG recordings in a number of neurodegenerative and neurodevelopmental conditions, these findings may be more broadly applicable in translational neuroscience.
Collapse
Affiliation(s)
- Jonathan W Lovelace
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
46
|
Pirbhoy PS, Rais M, Lovelace JW, Woodard W, Razak KA, Binder DK, Ethell IM. Acute pharmacological inhibition of matrix metalloproteinase-9 activity during development restores perineuronal net formation and normalizes auditory processing in Fmr1 KO mice. J Neurochem 2020; 155:538-558. [PMID: 32374912 DOI: 10.1111/jnc.15037] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
Individuals with Fragile X Syndrome (FXS) and autism spectrum disorder (ASD) exhibit cognitive impairments, social deficits, increased anxiety, and sensory hyperexcitability. Previously, we showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to abnormal development of parvalbumin (PV) interneurons and perineuronal nets (PNNs) in the developing auditory cortex (AC) of Fmr1 knock-out (KO) mice, which likely underlie auditory hypersensitivity. Thus, MMP-9 may serve as a potential target for treatment of auditory hypersensitivity in FXS. Here, we used the MMP-2/9 inhibitor, SB-3CT, to pharmacologically inhibit MMP-9 activity during a specific developmental period and to test whether inhibition of MMP-9 activity reverses neural oscillation deficits and behavioral impairments by enhancing PNN formation around PV cells in Fmr1 KO mice. Electroencephalography (EEG) was used to measure resting state and sound-evoked electrocortical activity in auditory and frontal cortices of postnatal day (P)22-23 male mice before and one-day after treatment with SB-3CT (25 mg/kg) or vehicle. At P27-28, animal behaviors were tested to measure the effects of the treatment on anxiety and hyperactivity. Results show that acute inhibition of MMP-9 activity improved evoked synchronization to auditory stimuli and ameliorated mouse behavioral deficits. MMP-9 inhibition enhanced PNN formation, increased PV levels and TrkB phosphorylation yet reduced Akt phosphorylation in the AC of Fmr1 KO mice. Our results show that MMP-9 inhibition during early postnatal development is beneficial in reducing some auditory processing deficits in the FXS mouse model and may serve as a candidate therapeutic for reversing sensory hypersensitivity in FXS and possibly other ASDs.
Collapse
Affiliation(s)
- Patricia S Pirbhoy
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Walker Woodard
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
47
|
Razak KA, Dominick KC, Erickson CA. Developmental studies in fragile X syndrome. J Neurodev Disord 2020; 12:13. [PMID: 32359368 PMCID: PMC7196229 DOI: 10.1186/s11689-020-09310-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/13/2020] [Indexed: 01/27/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of autism and intellectual disabilities. Humans with FXS exhibit increased anxiety, sensory hypersensitivity, seizures, repetitive behaviors, cognitive inflexibility, and social behavioral impairments. The main purpose of this review is to summarize developmental studies of FXS in humans and in the mouse model, the Fmr1 knockout mouse. The literature presents considerable evidence that a number of early developmental deficits can be identified and that these early deficits chart a course of altered developmental experience leading to symptoms well characterized in adolescents and adults. Nevertheless, a number of critical issues remain unclear or untested regarding the development of symptomology and underlying mechanisms. First, what is the role of FMRP, the protein product of Fmr1 gene, during different developmental ages? Does the absence of FMRP during early development lead to irreversible changes, or could reintroduction of FMRP or therapeutics aimed at FMRP-interacting proteins/pathways hold promise when provided in adults? These questions have implications for clinical trial designs in terms of optimal treatment windows, but few studies have systematically addressed these issues in preclinical and clinical work. Published studies also point to complex trajectories of symptom development, leading to the conclusion that single developmental time point studies are unlikely to disambiguate effects of genetic mutation from effects of altered developmental experience and compensatory plasticity. We conclude by suggesting a number of experiments needed to address these major gaps in the field.
Collapse
Affiliation(s)
- Khaleel A Razak
- Department of Psychology and Graduate Neuroscience Program, University of California, Riverside, USA
| | - Kelli C Dominick
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA
| | - Craig A Erickson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA. .,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA.
| |
Collapse
|
48
|
Nguyen AO, Binder DK, Ethell IM, Razak KA. Abnormal development of auditory responses in the inferior colliculus of a mouse model of Fragile X Syndrome. J Neurophysiol 2020; 123:2101-2121. [PMID: 32319849 DOI: 10.1152/jn.00706.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Sensory processing abnormalities are frequently associated with autism spectrum disorders, but the underlying mechanisms are unclear. Here we studied auditory processing in a mouse model of Fragile X Syndrome (FXS), a leading known genetic cause of autism and intellectual disability. Both humans with FXS and the Fragile X mental retardation gene (Fmr1) knockout (KO) mouse model show auditory hypersensitivity, with the latter showing a strong propensity for audiogenic seizures (AGS) early in development. Because midbrain abnormalities cause AGS, we investigated whether the inferior colliculus (IC) of the Fmr1 KO mice shows abnormal auditory processing compared with wild-type (WT) controls at specific developmental time points. Using antibodies against neural activity marker c-Fos, we found increased density of c-Fos+ neurons in the IC, but not auditory cortex, of Fmr1 KO mice at P21 and P34 following sound presentation. In vivo single-unit recordings showed that IC neurons of Fmr1 KO mice are hyperresponsive to tone bursts and amplitude-modulated tones during development and show broader frequency tuning curves. There were no differences in rate-level responses or phase locking to amplitude-modulated tones in IC neurons between genotypes. Taken together, these data provide evidence for the development of auditory hyperresponsiveness in the IC of Fmr1 KO mice. Although most human and mouse work in autism and sensory processing has centered on the forebrain, our new findings, along with recent work on the lower brainstem, suggest that abnormal subcortical responses may underlie auditory hypersensitivity in autism spectrum disorders.NEW & NOTEWORTHY Autism spectrum disorders (ASD) are commonly associated with sensory sensitivity issues, but the underlying mechanisms are unclear. This study presents novel evidence for neural correlates of auditory hypersensitivity in the developing inferior colliculus (IC) in Fmr1 knockout (KO) mouse, a mouse model of Fragile X Syndrome (FXS), a leading genetic cause of ASD. Responses begin to show genotype differences between postnatal days 14 and 21, suggesting an early developmental treatment window.
Collapse
Affiliation(s)
- Anna O Nguyen
- Bioengineering Program, University of California, Riverside, California
| | - Devin K Binder
- Graduate Neuroscience Program, University of California, Riverside, California.,Division of Biomedical Sciences, University of California, Riverside, California
| | - Iryna M Ethell
- Graduate Neuroscience Program, University of California, Riverside, California.,Division of Biomedical Sciences, University of California, Riverside, California
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, California.,Psychology Department, University of California, Riverside, California
| |
Collapse
|
49
|
McCullagh EA, Rotschafer SE, Auerbach BD, Klug A, Kaczmarek LK, Cramer KS, Kulesza RJ, Razak KA, Lovelace JW, Lu Y, Koch U, Wang Y. Mechanisms underlying auditory processing deficits in Fragile X syndrome. FASEB J 2020; 34:3501-3518. [PMID: 32039504 DOI: 10.1096/fj.201902435r] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 01/18/2020] [Indexed: 01/14/2023]
Abstract
Autism spectrum disorders (ASD) are strongly associated with auditory hypersensitivity or hyperacusis (difficulty tolerating sounds). Fragile X syndrome (FXS), the most common monogenetic cause of ASD, has emerged as a powerful gateway for exploring underlying mechanisms of hyperacusis and auditory dysfunction in ASD. This review discusses examples of disruption of the auditory pathways in FXS at molecular, synaptic, and circuit levels in animal models as well as in FXS individuals. These examples highlight the involvement of multiple mechanisms, from aberrant synaptic development and ion channel deregulation of auditory brainstem circuits, to impaired neuronal plasticity and network hyperexcitability in the auditory cortex. Though a relatively new area of research, recent discoveries have increased interest in auditory dysfunction and mechanisms underlying hyperacusis in this disorder. This rapidly growing body of data has yielded novel research directions addressing critical questions regarding the timing and possible outcomes of human therapies for auditory dysfunction in ASD.
Collapse
Affiliation(s)
- Elizabeth A McCullagh
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA.,Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
| | - Sarah E Rotschafer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Benjamin D Auerbach
- Center for Hearing and Deafness, Department of Communicative Disorders & Sciences, SUNY at Buffalo, Buffalo, NY, USA
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA
| | - Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, CA, USA
| | | | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ursula Koch
- Institute of Biology, Neurophysiology, Freie Universität Berlin, Berlin, Germany
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
50
|
Jonak CR, Lovelace JW, Ethell IM, Razak KA, Binder DK. Multielectrode array analysis of EEG biomarkers in a mouse model of Fragile X Syndrome. Neurobiol Dis 2020; 138:104794. [PMID: 32036032 DOI: 10.1016/j.nbd.2020.104794] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced inter-trial phase coherence of sound-evoked gamma oscillations. Identification of comparable EEG biomarkers in mouse models of FXS could facilitate the pre-clinical to clinical therapeutic pipeline. However, while human EEG studies have involved 128-channel scalp EEG acquisition, no mouse studies have been performed with more than three EEG channels. In the current study, we employed a recently developed 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in WT vs. Fmr1 KO mice. Using this system, we now report robust MEA-derived phenotypes including higher resting EEG power, altered event-related potentials (ERPs) and reduced inter-trial phase coherence to auditory chirp stimuli in Fmr1 KO mice that are remarkably similar to those reported in humans with FXS. We propose that the MEA system can be used for: (i) derivation of higher-level EEG parameters; (ii) EEG biomarkers for drug testing; and (ii) mechanistic studies of FXS pathophysiology.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America
| | - Jonathan W Lovelace
- Department of Psychology, University of California, Riverside, United States of America
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, United States of America; Department of Psychology, University of California, Riverside, United States of America
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America.
| |
Collapse
|