1
|
Li R, Anzai M, Shibata A, Ito-Ishida A. Synaptic disturbance in neurodevelopmental disorders: Perspectives from fragile X and Rett syndromes. Brain Dev 2025; 47:104358. [PMID: 40228442 DOI: 10.1016/j.braindev.2025.104358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
Neurodevelopmental disorders (NDDs) are often referred to as "synaptopathies" because many of their behavioral symptoms arise from impaired synaptic development and function. However, the mechanisms that connect synaptic dysfunction to neurological symptoms remain unclear, mainly due to the wide variety of genetic and environmental factors involved in these disorders. Fragile X syndrome and Rett syndrome, two extensively studied monogenic NDDs, provide a unique opportunity to explore these mechanisms at molecular, cellular, and synaptic levels. This review summarizes the current understanding of how synaptic alterations contribute to the neurological symptoms observed in fragile X and Rett syndromes. A comparison of findings from mouse models indicates that an imbalance in local and distal connectivity may serve as a common feature of both disorders.
Collapse
Affiliation(s)
- Ruixiang Li
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan; Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Mai Anzai
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Akiko Shibata
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Aya Ito-Ishida
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
2
|
Zheng S, Chen C. Auditory processing deficits in autism spectrum disorder: mechanisms, animal models, and therapeutic directions. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02919-x. [PMID: 40353881 DOI: 10.1007/s00702-025-02919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025]
Abstract
Auditory processing abnormalities are a prominent feature of Autism Spectrum Disorder (ASD), significantly affecting sensory integration, communication, and social interaction. This review delves into the neurobiological mechanisms underlying these deficits, including structural and functional disruptions in the auditory cortex, imbalances in excitatory and inhibitory signaling, and synaptic dysfunction. Genetic contributions from mutations in CNTNAP2, SHANK3, FMR1, and FOXP2 are explored, highlighting their roles in auditory abnormalities. Animal models, such as BTBRT+tf/J mice (BTBR) and valproic acid (VPA)-exposed rodents, provide critical insights into the sensory abnormalities observed in ASD. In addition, the review discusses current pharmacological strategies and emerging interventions targeting neurotransmitter systems and synaptic plasticity. Notably, future directions are emphasized, highlighting the need for integrated pharmacological and auditory-specific therapies to enhance sensory processing and communication outcomes in ASD. Overall, this review aims to bridge the gap between basic neurobiological research and clinical application, guiding future studies and therapeutic developments in ASD-related auditory processing deficits.
Collapse
Affiliation(s)
- Shuyu Zheng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310051, Zhejiang, China
- Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Chen Chen
- Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
3
|
Singh A, Rizzi M, Seo SS, Osterweil EK. Syngap+/- CA1 Pyramidal Neurons Exhibit Upregulated Translation of Long MRNAs Associated with LTP. eNeuro 2025; 12:ENEURO.0086-25.2025. [PMID: 40295099 PMCID: PMC12091090 DOI: 10.1523/eneuro.0086-25.2025] [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: 02/19/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
In the Syngap+/- model of SYNGAP1-related intellectual disability (SRID), excessive neuronal protein synthesis is linked to deficits in synaptic plasticity. Here, we use Translating Ribosome Affinity Purification and RNA-seq (TRAP-seq) to identify mistranslating mRNAs in Syngap+/- CA1 pyramidal neurons that exhibit occluded long-term potentiation (LTP). We find the translation environment is significantly altered in a manner that is distinct from the Fmr1-/y model of fragile X syndrome (FXS), another monogenic model of autism and intellectual disability. The Syngap+/- translatome is enriched for regulators of DNA repair and mimics changes induced with chemical LTP (cLTP) in WT. This includes a striking upregulation in the translation of mRNAs with a longer-length (>2 kb) coding sequence (CDS). In contrast, long CDS transcripts are downregulated with induction of Gp1 metabotropic glutamate receptor-induced long-term depression (mGluR-LTD) in WT, and in the Fmr1-/y model that exhibits occluded mGluR-LTD. Together, our results show the Syngap+/- and Fmr1-/y models mimic the translation environments of LTP and LTD, respectively, consistent with the saturation of plasticity states in each model. Moreover, we show that translation of >2 kb mRNAs is a defining feature of LTP that is oppositely regulated during LTD, revealing a novel mRNA signature of plasticity.
Collapse
Affiliation(s)
- Aditi Singh
- Rosamund Stone Zander Translational Neuroscience Center, F. M. Kirby Center, Department of Neurology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Manuela Rizzi
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Sang S Seo
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Emily K Osterweil
- Rosamund Stone Zander Translational Neuroscience Center, F. M. Kirby Center, Department of Neurology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
4
|
D'Addario SL, Rosina E, Massaro Cenere M, Bagni C, Mercuri NB, Ledonne A. ErbB inhibition rescues nigral dopamine neuron hyperactivity and repetitive behaviors in a mouse model of fragile X syndrome. Mol Psychiatry 2025; 30:2183-2196. [PMID: 39543371 PMCID: PMC12014506 DOI: 10.1038/s41380-024-02831-y] [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: 04/16/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Repetitive stereotyped behaviors are core symptoms of autism spectrum disorders (ASD) and fragile X syndrome (FXS), the prevalent genetic cause of intellectual disability and autism. The nigrostriatal dopamine (DA) circuit rules movement and creation of habits and sequential behaviors; therefore, its dysregulation could promote autistic repetitive behaviors. Nevertheless, inspection of substantia nigra pars compacta (SNpc) DA neurons in ASD models has been overlooked and specific evidence of their altered activity in ASD and FXS is absent. Here, we show that hyperactivity of SNpc DA neurons is an early feature of FXS. The underlying mechanism relies on an interplay between metabotropic glutamate receptor 1 (mGluR1) and ErbB tyrosine kinases, receptors for the neurotrophic and differentiation factors known as neuregulins. Up-regulation of ErbB4 and ErbB2 in nigral DA neurons drives neuronal hyperactivity and repetitive behaviors of the FXS mouse, concurrently rescued by ErbB inhibition. In conclusion, beyond providing the first evidence that nigral DA neuron hyperactivity is a signature of FXS and nigral mGluR1 and ErbB4/2 play a relevant role in FXS etiology, we demonstrate that inhibiting ErbB is a valuable pharmacological approach to attenuate stereotyped repetitive behaviors, thus opening an avenue toward innovative therapies for ASD and FXS treatment.
Collapse
Affiliation(s)
| | - Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Neurology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Pharmacology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
5
|
Bonnycastle K, Nawaz MS, Kind PC, Cousin MA. Convergent depression of activity-dependent bulk endocytosis in rodent models of autism spectrum disorder. Mol Autism 2025; 16:26. [PMID: 40241211 PMCID: PMC12004638 DOI: 10.1186/s13229-025-00660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The key pathological mechanisms underlying autism spectrum disorder (ASD) remain relatively undetermined, potentially due to the heterogenous nature of the condition. Targeted studies of a series of monogenic ASDs have revealed postsynaptic dysfunction as a central conserved mechanism. Presynaptic dysfunction is emerging as an additional disease locus in neurodevelopmental disorders; however, it is unclear whether this dysfunction drives ASD or is an adaptation to the altered brain microenvironment. METHODS To differentiate between these two competing scenarios, we performed a high content analysis of key stages of the synaptic vesicle lifecycle in primary neuronal cultures derived from a series of preclinical rat models of monogenic ASD. These five independent models (Nrxn1+/-, Nlgn3-/y, Syngap+/-, Syngap+/Δ-GAP, Pten+/-) were specifically selected to have perturbations in a diverse palette of genes that were expressed either at the pre- or post-synapse. Synaptic vesicle exocytosis and cargo trafficking were triggered via two discrete trains of activity and monitored using the genetically-encoded reporter synaptophysin-pHluorin. Activity-dependent bulk endocytosis was assessed during intense neuronal activity using the fluid phase marker tetramethylrhodamine-dextran. RESULTS Both synaptic vesicle fusion events and cargo trafficking were unaffected in all models investigated under all stimulation protocols. However, a key convergent phenotype across neurons derived from all five models was revealed, a depression in activity-dependent bulk endocytosis. LIMITATIONS The study is exclusively conducted in primary cultures of hippocampal neurons; therefore, the impact on neurons from other brain regions or altered brain microcircuitry was not assessed. No molecular mechanism has been identified for this depression. CONCLUSION This suggests that depression of activity-dependent bulk endocytosis is a presynaptic homeostatic mechanism to correct for intrinsic dysfunction in ASD neurons.
Collapse
Affiliation(s)
- Katherine Bonnycastle
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Mohammed Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
| |
Collapse
|
6
|
Shi Z, Wen K, Sammudin NH, LoRocco N, Zhuang X. Erasing "bad memories": reversing aberrant synaptic plasticity as therapy for neurological and psychiatric disorders. Mol Psychiatry 2025:10.1038/s41380-025-03013-0. [PMID: 40210977 DOI: 10.1038/s41380-025-03013-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/24/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
Dopamine modulates corticostriatal plasticity in both the direct and indirect pathways of the cortico-striato-thalamo-cortical (CSTC) loops. These gradual changes in corticostriatal synaptic strengths produce long-lasting changes in behavioral responses. Under normal conditions, these mechanisms enable the selection of the most appropriate responses while inhibiting others. However, under dysregulated dopamine conditions, including a lack of dopamine release or dopamine signaling, these mechanisms could lead to the selection of maladaptive responses and/or the inhibition of appropriate responses in an experience-dependent and task-specific manner. In this review, we propose that preventing or reversing such maladaptive synaptic strengths and erasing such aberrant "memories" could be a disease-modifying therapeutic strategy for many neurological and psychiatric disorders. We review evidence from Parkinson's disease, drug-induced parkinsonism, L-DOPA-induced dyskinesia, obsessive-compulsive disorder, substance use disorders, and depression as well as research findings on animal disease models. Altogether, these studies allude to an emerging theme in translational neuroscience and promising new directions for therapy development. Specifically, we propose that combining pharmacotherapy with behavioral therapy or with deep brain stimulation (DBS) could potentially cause desired changes in specific neural circuits. If successful, one important advantage of correcting aberrant synaptic plasticity is long-lasting therapeutic effects even after treatment has ended. We will also discuss the potential molecular targets for these therapeutic approaches, including the cAMP pathway, proteins involved in synaptic plasticity as well as pathways involved in new protein synthesis. We place special emphasis on RNA binding proteins and epitranscriptomic mechanisms, as they represent a new frontier with the distinct advantage of rapidly and simultaneously altering the synthesis of many proteins locally.
Collapse
Affiliation(s)
- Zhuoyue Shi
- The Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Kailong Wen
- The Committee on Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Nabilah H Sammudin
- The Committee on Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Nicholas LoRocco
- The Interdisciplinary Scientist Training Program, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoxi Zhuang
- The Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA.
- The Neuroscience Institute, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
7
|
Lakhani A, Huang W, Rodgers CC, Wenner P. Impairment in the homeostatic recruitment of layer 5/6 neurons following whisker stimulation in Fmr1 KO mice. Neurobiol Dis 2025; 207:106837. [PMID: 39938578 PMCID: PMC12019994 DOI: 10.1016/j.nbd.2025.106837] [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: 10/14/2024] [Revised: 02/08/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025] Open
Abstract
Mouse models of Fragile X Syndrome (FXS) have demonstrated impairments in sensory-evoked neuronal firing of excitatory and inhibitory neurons. Homeostatic plasticity does not compensate for these changes in activity. Previous work has shown that impairments in homeostatic plasticity mechanisms are observed in FXS, including deficits in synaptic scaling and intrinsic excitability. Here, we aimed to examine how sensory integration changes in vivo following a homeostatic perturbation, unilateral whisker deprivation (WD), in an Fmr1 knock out (KO) mouse model. We used multi-electrode array recordings of neurons in the lightly anesthetized juvenile mouse somatosensory cortex, and found that whisker-evoked responses in layer 5/6 (L5/6) excitatory neurons were weaker in the KO compared to the wild-type (WT). We show that WD in the WT leads to a compensatory increase in the proportion of L5/6 somatosensory neurons that were recruited following whisker stimulation, but this did not occur in the KO. On the other hand, certain compensatory responses were observed in the KO following WD; the firing rate of the whisker-responsive neurons was increased following both a 2- and 7-day WD. Similar to excitatory neurons, we observed increased recruitment of fast spiking (presumed inhibitory) neurons following WD in the WT, but not KO. Our results suggest that certain homeostatic mechanisms are impaired in the KO, while others appear to remain intact. Compromised homeostatic plasticity in development could influence adult sensory processing and long-term cortical organization.
Collapse
Affiliation(s)
- Alishah Lakhani
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States of America.
| | - Washington Huang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Chris C Rodgers
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, United States of America.
| | - Peter Wenner
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, United States of America.
| |
Collapse
|
8
|
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
|
9
|
Westmark CJ. Soy-based purified ingredient diet affects mouse gut permeability and the microbiome in fragile X mice. Front Mol Neurosci 2025; 18:1520211. [PMID: 40190341 PMCID: PMC11968763 DOI: 10.3389/fnmol.2025.1520211] [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: 10/30/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Gastrointestinal problems including vomiting, reflux, flatulence, diarrhea, constipation and colic are common comorbidities in fragile X syndrome. There is accumulating evidence suggesting that leaky gut syndrome causes neurological phenotypes. Although fragile X messenger ribonucleoprotein is ubiquitously expressed, there is a dearth of knowledge regarding its role outside of the brain including effects on gut dysfunction in fragile X. The aim of this study was to generate novel data on gastrointestinal barrier function and the gut microbiome in response to Fmr1 genotype, sex and diet in mice. Methods Fmr1KO male mice and littermate controls in an FVB background were maintained on two purified ingredient diets (AIN-93G with casein protein versus soy protein isolate) versus two standard chows (Teklad 2019 with wheat, corn and yeast protein versus Purina 5015 with wheat, soy, corn, yeast and whey protein sources). Gut permeability was quantified by FITC-dextran levels in blood plasma. The cecal microbiome was identified by 16S rRNA sequencing. In addition, gut permeability was tested in Fmr1KO mice in the C57BL/6 J background maintained on casein- and soy protein isolate-based AIN-93G versus Teklad 2019. Results Knockout of the Fmr1 gene in FVB mice did not affect gut permeability. Soy protein isolate-based AIN-93G increased gut permeability. Beta-diversity of the cecal microbiome was significantly altered as a function of the four test diets. Akkermansia_muciniphila was increased in Fmr1KO mice fed AIN-93G while unnamed species within the genus Anaerovorax and family Ruminococcaceae were increased and the order Clostridales decreased in Fmr1KO mice fed AIN-93G/soy. Fmr1KO mice in the C57BL/6 J background exhibited increased gut permeability in response to soy protein. Discussion These findings regarding the effects of diet on gut permeability and the microbiome have important implications for experimental design. Single-source diets are ubiquitously used to maintain laboratory animals for medical research and feed details are frequently not reported in publications. Diet/phenotype interactions could have a large impact on inter-laboratory replicability in premedical research. For infants with fragile X, early-life diet could impact the severity of disease outcomes.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
10
|
Ciranna L, Costa L. Therapeutic Effects of Pharmacological Modulation of Serotonin Brain System in Human Patients and Animal Models of Fragile X Syndrome. Int J Mol Sci 2025; 26:2495. [PMID: 40141138 PMCID: PMC11941774 DOI: 10.3390/ijms26062495] [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: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
The brain serotonin (5-HT) system modulates glutamatergic and GABAergic transmission in almost every brain area, crucially regulating mood, food intake, body temperature, pain, hormone secretion, learning and memory. Previous studies suggest a disruption of the brain 5-HT system in Fragile X Syndrome, with abnormal activity of the 5-HT transporter leading to altered 5-HT brain levels. We provide an update on therapeutic effects exerted by drugs modulating serotonergic transmission on Fragile X patients and animal models. The enhancement of serotonergic transmission using Selective Serotonin Reuptake Inhibitors (SSRIs) corrected mood disorders and language deficits in Fragile X patients. In Fmr1 KO mice, a model of Fragile X Syndrome, selective 5-HT7 receptor agonists rescued synaptic plasticity, memory and stereotyped behavior. In addition, drugs specifically acting on 5-HT1A, 5-HT2 and 5-HT5 receptor subtypes were able to correct, respectively, epilepsy, learning deficits and hyperactivity in different Fragile X animal models. In conclusion, the SSRI treatment of Fragile X patients improves mood and language; in parallel, studies on animal models suggest that compounds selectively acting on distinct 5-HT receptor subtypes might provide a targeted correction of other Fragile X phenotypes, and thus should be further tested in clinical trials for future therapy.
Collapse
Affiliation(s)
- Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
11
|
Lau JCY, Guilfoyle J, Crawford S, Johnson G, Landau E, Xing J, Kumareswaran M, Ethridge S, Butler M, Goldman L, Martin GE, Zhou L, Krizman J, Nicol T, Kraus N, Berry-Kravis E, Losh M. Prosodic Differences in Women with the FMR1 Premutation: Subtle Expression of Autism-Related Phenotypes Through Speech. Int J Mol Sci 2025; 26:2481. [PMID: 40141125 PMCID: PMC11942500 DOI: 10.3390/ijms26062481] [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: 02/08/2025] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Evidence suggests that carriers of FMR1 mutations (e.g., fragile X syndrome and the FMR1 premutation) may demonstrate specific phenotypic patterns shared with autism (AU), particularly in the domain of pragmatic language, which involves the use of language in social contexts. Such evidence may implicate FMR1, a high-confidence gene associated with AU, in components of the AU phenotype. Prosody (i.e., using intonation and rhythm in speech to express meaning) is a pragmatic feature widely impacted in AU. Prosodic differences have also been observed in unaffected relatives of autistic individuals and in those with fragile X syndrome, although prosody has not been extensively studied among FMR1 premutation carriers. This study investigated how FMR1 variability may specifically influence prosody by examining the prosodic characteristics and related neural processing of prosodic features in women carrying the FMR1 premutation (PM). In Study 1, acoustic measures of prosody (i.e., in intonation and rhythm) were examined in speech samples elicited from a semi-structured narrative task. Study 2 examined the neural frequency following response (FFR) as an index of speech prosodic processing. Findings revealed differences in the production of intonation and rhythm in PM carriers relative to controls, with patterns that parallel differences identified in parents of autistic individuals. No differences in neural processing of prosodic cues were found. Post hoc analyses further revealed associations between speech rhythm and FMR1 variation (number of CGG repeats) among PM carriers. Together, the results suggest that FMR1 may play a role in speech prosodic phenotypes, at least in speech production, contributing to a deeper understanding of AU-related speech and language phenotypes among FMR1 mutation carriers.
Collapse
Affiliation(s)
- Joseph C. Y. Lau
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Janna Guilfoyle
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Stephanie Crawford
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Grace Johnson
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Emily Landau
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Jiayin Xing
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Mitra Kumareswaran
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Sarah Ethridge
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Maureen Butler
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Lindsay Goldman
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Gary E. Martin
- Department of Communication Sciences and Disorders, St. John’s University, Queens, NY 11439, USA;
| | - Lili Zhou
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA; (L.Z.); (E.B.-K.)
| | - Jennifer Krizman
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Trent Nicol
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Nina Kraus
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA; (L.Z.); (E.B.-K.)
| | - Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA; (J.C.Y.L.); (J.G.); (S.C.); (G.J.); (E.L.); (J.X.); (M.K.); (S.E.); (M.B.); (L.G.); (J.K.); (T.N.); (N.K.)
| |
Collapse
|
12
|
Wang C, Liu J, Su L, Wang X, Bian Y, Wang Z, Ye L, Lu X, Zhou L, Chen W, Yang W, Liu J, Wang L, Shen Y. GABAergic Progenitor Cell Graft Rescues Cognitive Deficits in Fragile X Syndrome Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411972. [PMID: 39823534 PMCID: PMC11904963 DOI: 10.1002/advs.202411972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Fragile X syndrome (FXS) is an inherited neurodevelopmental disorder characterized by a range of clinical manifestations with no effective treatment strategy to date. Here, transplantation of GABAergic precursor cells from the medial ganglionic eminence (MGE) is demonstrated to significantly improve cognitive performance in Fmr1 knockout (KO) mice. Within the hippocampus of Fmr1-KO mice, MGE-derived cells from wild-type donor mice survive, migrate, differentiate into functionally mature interneurons, and form inhibitory synaptic connections with host pyramidal neurons. MGE cell transplantation restores Ras-PKB signaling in pyramidal neurons, enhances AMPA receptor trafficking, rescues synaptic plasticity, and corrects abnormal hippocampal neural oscillations. These findings highlight the potential of GABAergic precursor cell transplantation as a promising therapeutic strategy for FXS.
Collapse
Affiliation(s)
- Chen Wang
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Jia‐Yu Liu
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Zhejiang Development & Planning InstituteHangzhou310030China
| | - Li‐Da Su
- Neuroscience Care UnitKey Laboratory of Multiple Organ Failure of Ministry of Educationthe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou310009China
- Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang ProvinceHangzhou310009China
| | - Xin‐Tai Wang
- Institute of Life SciencesCollege of Life and Environmental SciencesHangzhou Normal UniversityHangzhou311121China
| | - Yu‐Peng Bian
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | | | - Lu‐Yu Ye
- Department of BiophysicsZhejiang University School of MedicineHangzhou310058China
| | - Xin‐Jiang Lu
- Department of PhysiologyZhejiang University School of MedicineHangzhou310058China
| | - Lin Zhou
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Wei Chen
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Wei Yang
- Department of BiophysicsZhejiang University School of MedicineHangzhou310058China
| | - Jun Liu
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
| | - Luxi Wang
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Ying Shen
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Key Laboratory for Precision DiagnosisTreatmentand Clinical Translation of Rare Diseases of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| |
Collapse
|
13
|
Huang L, Zhao B, Wan Y. Disruption of RNA-binding proteins in neurological disorders. Exp Neurol 2025; 385:115119. [PMID: 39709152 DOI: 10.1016/j.expneurol.2024.115119] [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: 09/09/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
RNA-binding proteins (RBPs) are multifunctional proteins essential for the regulation of RNA processing and metabolism, contributing to the maintenance of cell homeostasis by modulating the expression of target genes. Many RBPs have been associated with neuron-specific processes vital for neuronal development and survival. RBP dysfunction may result in aberrations in RNA processing, which subsequently initiate a cascade of effects. Notably, RBPs are involved in the onset and progression of neurological disorders via diverse mechanisms. Disruption of RBPs not only affects RNA processing, but also promotes the abnormal aggregation of proteins into toxic inclusion bodies, and contributes to immune responses that drive the progression of neurological diseases. In this review, we summarize recent discoveries relating to the roles of RBPs in neurological diseases, discuss their contributions to such conditions, and highlight the unique functions of these RBPs within the nervous system.
Collapse
Affiliation(s)
- Luyang Huang
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China
| | - Bo Zhao
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China
| | - Youzhong Wan
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China.
| |
Collapse
|
14
|
Lu L, Sarkar AK, Dao L, Liu Y, Ma C, Thwin PH, Chang X, Yoshida G, Li A, Wang C, Westerkamp C, Schmitt L, Chelsey M, Stephanie M, Zhao Y, Liu Y, Wang X, Zhu LQ, Liu D, Tchieu J, Miyakoshi M, Zhu H, Gross C, Pedapati E, Salomonis N, Erickson C, Guo Z. An iPSC model of fragile X syndrome reflects clinical phenotypes and reveals m 6 A- mediated epi-transcriptomic dysregulation underlying synaptic dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.14.618205. [PMID: 39464060 PMCID: PMC11507714 DOI: 10.1101/2024.10.14.618205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Fragile X syndrome (FXS), the leading genetic cause of intellectual disability, arises from FMR1 gene silencing and loss of the FMRP protein. N6-methyladenosine (m 6 A) is a prevalent mRNA modification essential for post-transcriptional regulation. FMRP is known to bind to and regulate the stability of m 6 A-containing transcripts. However, how loss of FMRP impacts on transcriptome-wide m 6 A modifications in FXS patients remains unknown. To answer this question, we generated cortical neurons differentiated from induced pluripotent stem cells (iPSC) derived from healthy subjects and FXS patients. In electrophysiology recordings, we validated that synaptic and neuronal network defects in iPSC-derived FXS neurons corresponded to the clinical EEG data of the patients from which the corresponding iPSC line was derived. In analysis of transcriptome-wide methylation, we show that FMRP deficiency led to increased translation of m 6 A writers, resulting in hypermethylation that primarily affecting synapse-associated transcripts and increased mRNA decay. Conversely, in the presence of an m 6 A writer inhibitor, synaptic defects in FXS neurons were rescued. Taken together, our findings uncover that an FMRP-dependent epi-transcriptomic mechanism contributes to FXS pathogenesis by disrupting m 6 A modifications in FXS, suggesting a promising avenue for m 6 A- targeted therapies.
Collapse
|
15
|
Singer RA, Rajchin V, Park K, Heintz N, Darnell RB. Opto-CLIP reveals dynamic FMRP regulation of mRNAs upon CA1 neuronal activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.13.607210. [PMID: 39185177 PMCID: PMC11343148 DOI: 10.1101/2024.08.13.607210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Neuronal diversity and function are intricately linked to the dynamic regulation of RNA metabolism. Electrophysiologic studies of synaptic plasticity, models for learning and memory, are disrupted in Fragile X Syndrome (FXS). FXS is characterized by the loss of FMRP, an RNA-binding protein (RBP) known to suppress translation of specific neuronal RNAs. Synaptic plasticity in CA1 excitatory hippocampal neurons is protein-synthesis dependent, suggesting a role for FMRP in FXS-related synaptic deficits. To explore this model, we developed Opto-CLIP, integrating optogenetics with cell-type specific FMRP-CLIP and RiboTag in CA1 neurons, allowing investigation of activity-induced FMRP regulation. We tracked changes in FMRP binding and ribosome-associated RNA profiles 30 minutes after neuronal activation. Our findings reveal distinct temporal dynamics for FMRP transcript regulation in the cell body versus the synapse. In the cell body, FMRP binding to transcripts encoding nuclear functions is relieved, potentially allowing rapid transcriptional responses to neuronal activation. At the synapse, FMRP binding to transcripts encoding synaptic targets was relatively stable, with variability in translational control across target categories. These results offer fresh insights into the dynamic regulation of RNA by FMRP in response to neuronal activation and provide a foundation for future research into the mechanisms of RBP-mediated synaptic plasticity.
Collapse
Affiliation(s)
- Ruth A. Singer
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Veronika Rajchin
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Kwanghoon Park
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Robert B. Darnell
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Lead Contact
| |
Collapse
|
16
|
Dionne O, Sabatié S, Laurent B. Deciphering the physiopathology of neurodevelopmental disorders using brain organoids. Brain 2025; 148:12-26. [PMID: 39222411 PMCID: PMC11706293 DOI: 10.1093/brain/awae281] [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: 03/30/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Neurodevelopmental disorders (NDD) encompass a range of conditions marked by abnormal brain development in conjunction with impaired cognitive, emotional and behavioural functions. Transgenic animal models, mainly rodents, traditionally served as key tools for deciphering the molecular mechanisms driving NDD physiopathology and significantly contributed to the development of pharmacological interventions aimed at treating these disorders. However, the efficacy of these treatments in humans has proven to be limited, due in part to the intrinsic constraint of animal models to recapitulate the complex development and structure of the human brain but also to the phenotypic heterogeneity found between affected individuals. Significant advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising avenue for overcoming these challenges. Indeed, the development of advanced differentiation protocols for generating iPSC-derived brain organoids gives an unprecedented opportunity to explore human neurodevelopment. This review provides an overview of how 3D brain organoids have been used to investigate various NDD (i.e. Fragile X syndrome, Rett syndrome, Angelman syndrome, microlissencephaly, Prader-Willi syndrome, Timothy syndrome, tuberous sclerosis syndrome) and elucidate their pathophysiology. We also discuss the benefits and limitations of employing such innovative 3D models compared to animal models and 2D cell culture systems in the realm of personalized medicine.
Collapse
Affiliation(s)
- Olivier Dionne
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Salomé Sabatié
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5H4, Canada
| |
Collapse
|
17
|
Janz P, Bainier M, Marashli S, Gross S, Redondo RL. Clinically-probed mechanisms of action in Fragile-X syndrome fail to normalize translational EEG phenotypes in Fmr1 knockout mice. Neuropharmacology 2025; 262:110182. [PMID: 39396738 DOI: 10.1016/j.neuropharm.2024.110182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by Fragile X Messenger Ribonucleoprotein (FMRP) deficiency. Electroencephalogram (EEG) changes in FXS include alterations of oscillatory activity and responses to sensory stimuli, some of which have been back-translated into rodent models by knocking-out the Fragile X messenger ribonucleoprotein 1 gene (Fmr1-KO). However, the validity of these EEG phenotypes as objective biomarkers requires further investigation. Potential pharmacotherapies such as mGluR5 inhibitors (e.g. CTEP; 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazole-4-yl)ethynyl)pyridine), GABABR agonists (e.g. arbaclofen) and δ-containing GABAAR agonists (e.g. gaboxadol) have not translated into clinical success despite rescuing many phenotypes in the Fmr1-KO model. Yet none of these treatments have been assessed on EEG phenotypes in the Fmr1-KO model. Therefore, we set out to discover new EEG phenotypes in Fmr1-KO mice, using "task-free" and auditory-evoked (AEPs) and visually-evoked potential (VEP) paradigms, and probe their modulation by CTEP, arbaclofen and gaboxadol, using within-subjects designs. First, we report Fmr1-KO-associated EEG abnormalities that closely resemble those observed in FXS, including elevated gamma-band power, reduced alpha/beta-band coherence, increased AEPs and delayed VEPs. Secondly, we found that pharmacological treatment, at best, only partially normalized EEG phenotypes. CTEP restored alpha/beta-band coherence and AEP amplitudes but failed to normalize gamma power and VEP latencies. Conversely, arbaclofen reduced gamma power but did not restore coherence or AEP amplitudes and further delayed VEPs. Gaboxadol did not normalize any EEG phenotypes. We conclude that these compounds have limited ability to normalize these EEG phenotypes.
Collapse
Affiliation(s)
- Philipp Janz
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Marie Bainier
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Samuel Marashli
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Simon Gross
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Roger L Redondo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| |
Collapse
|
18
|
Fink JJ, Delaney-Busch N, Dawes R, Nanou E, Folts C, Harikrishnan K, Hempel C, Upadhyay H, Nguyen T, Shroff H, Stoppel D, Ryan SJ, Jacques J, Grooms J, Berry-Kravis E, Bear MF, Williams LA, Gerber D, Bunnage M, Furey B, Dempsey GT. Deep functional measurements of Fragile X syndrome human neurons reveal multiparametric electrophysiological disease phenotype. Commun Biol 2024; 7:1447. [PMID: 39506078 PMCID: PMC11541539 DOI: 10.1038/s42003-024-07120-6] [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: 10/12/2023] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by hypermethylation of expanded CGG repeats (>200) in the FMR1 gene leading to gene silencing and loss of Fragile X Messenger Ribonucleoprotein (FMRP) expression. FMRP plays important roles in neuronal function, and loss of FMRP in mouse and human FXS cell models leads to aberrant synaptic signaling and hyperexcitability. Multiple drug candidates have advanced into clinical trials for FXS, but no efficacious treatment has been identified to date, possibly as a consequence of poor translation from pre-clinical animal models to human. Here, we use a high resolution all-optical electrophysiology platform applied to multiple FXS patient-derived and CRISPR/Cas9-generated isogenic neuronal cell lines to develop a multi-parametric FXS disease phenotype. This neurophysiological phenotype was optimized and validated into a high throughput assay based on the amount of FMRP re-expression and the number of healthy neurons in a mosaic network necessary for functional rescue. The resulting highly sensitive and multiparameter functional assay can now be applied as a discovery platform to explore new therapeutic approaches for the treatment of FXS.
Collapse
Affiliation(s)
- James J Fink
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | - David Stoppel
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven J Ryan
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | - Jane Jacques
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | - Jennifer Grooms
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | | | - Mark F Bear
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luis A Williams
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | - David Gerber
- Quiver Bioscience, Cambridge, MA, USA
- Q-State Biosciences, Cambridge, MA, USA
| | | | | | - Graham T Dempsey
- Quiver Bioscience, Cambridge, MA, USA.
- Q-State Biosciences, Cambridge, MA, USA.
| |
Collapse
|
19
|
Napier M, Kumar A, Szulist N, Martin D, Scott AL. P2X7 expression patterns in the developing Fmr1-knockout mouse hippocampus. Hippocampus 2024; 34:633-644. [PMID: 39269925 DOI: 10.1002/hipo.23634] [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: 12/22/2023] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
Fragile-X Syndrome (FXS) is the leading monogenetic cause of intellectual disability among children but remains without a cure. Using the Fmr1 KO mouse model of FXS, much work has been done to understand FXS hippocampus dysfunction. Purinergic signaling, where ATP and its metabolites are used as signaling molecules, participates in hippocampus development, but it is unknown if purinergic signaling is affected in the developing Fmr1 KO hippocampus. In our study, we characterized the purinergic receptor P2X7. We first found that P2X7 was reduced in Fmr1 KO whole hippocampus tissue at P14 and P21, corresponding to the periods of neurite outgrowth and synaptic refinement in the hippocampus. We then evaluated the cell-specific expression of P2X7 with immunofluorescence and found differences between WT and Fmr1 KO mice in P2X7 colocalization with hippocampal microglia and neurons. P2X7 colocalized more with microglia at P14 and P21, but there was a sex-specific reduction in P2X7 colocalization with neurons. In contrast, male mice at P14 and P21 showed reduced neuronal P2X7 colocalization compared to females, but only females showed reduced absolute neuronal P2X7 expression across the dorsal hippocampal formation. Together, our results suggest that P2X7 expression is altered during Fmr1-KO hippocampal development, potentially influencing several developmental processes in the Fmr1-KO hippocampus formation.
Collapse
Affiliation(s)
- Matthew Napier
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Ashish Kumar
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Natasha Szulist
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dale Martin
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Angela L Scott
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
20
|
Yang Y, Chen BR, Ye XC, Ni LY, Zhang XY, Liu YZ, Lyu TJ, Tian Y, Fu YJ, Wang Y. The chromodomain protein CDYL confers forebrain identity to human cortical organoids by inhibiting neuronatin. Cell Rep 2024; 43:114814. [PMID: 39378153 DOI: 10.1016/j.celrep.2024.114814] [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: 12/01/2023] [Revised: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
Fate determination of neural stem cells (NSCs) is crucial for cortex development and is closely linked to neurodevelopmental disorders when gene expression networks are disrupted. The transcriptional corepressor chromodomain Y-like (CDYL) is widely expressed across diverse cell populations within the human embryonic cortex. However, its precise role in cortical development remains unclear. Here, we show that CDYL is critical for human cortical neurogenesis and that its deficiency leads to a substantial increase in gamma-aminobutyric acid (GABA)-ergic neurons in cortical organoids. Subsequently, neuronatin (NNAT) is identified as a significant target of CDYL, and its abnormal expression obviously influences the fate commitment of cortical NSCs. Cross-species comparisons of CDYL targets unravel a distinct developmental trajectory between human cortical organoids and the mouse cortex at an analogous stage. Collectively, our data provide insight into the evolutionary roles of CDYL in human cortex development, emphasizing its critical function in maintaining the fate of human cortical NSCs.
Collapse
Affiliation(s)
- Yaming Yang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Bai-Rong Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xi-Chun Ye
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Liang-Yu Ni
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xi-Yin Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun-Ze Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Tian-Jie Lyu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Yue Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun-Jie Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
21
|
Protic D, Breeze E, Mendoza G, Zafarullah M, Abbeduto L, Hagerman R, Coffey C, Cudkowicz M, Durbin-Johnson B, Ashwood P, Berry-Kravis E, Erickson CA, Filipink R, Gropman A, Lehwald L, Maxwell-Horn A, Morris S, Bennett AP, Prock L, Talboy A, Tartaglia N, Veenstra-VanderWeele J, Tassone F. Negative effect of treatment with mGluR5 negative allosteric modulator AFQ056 on blood biomarkers in young individuals with Fragile X syndrome. SAGE Open Med 2024; 12:20503121241282401. [PMID: 39483619 PMCID: PMC11526204 DOI: 10.1177/20503121241282401] [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: 04/30/2024] [Accepted: 08/22/2024] [Indexed: 11/03/2024] Open
Abstract
Background Fragile X syndrome, with an approximate incidence rate of 1 in 4000 males to 1 in 8000 females, is the most prevalent genetic cause of heritable intellectual disability and the most common monogenic cause of autism spectrum disorder. The full mutation of the Fragile X Messenger Ribonucleoprotein-1 gene, characterized by an expansion of CGG trinucleotide repeats (>200 CGG repeats), leads to fragile X syndrome. Currently, there are no targeted treatments available for fragile X syndrome. In a recent large multi-site trial, FXLEARN, the effects of the mGluR5 negative allosteric modulator, AFQ056 (mavoglurant), were investigated, but did not show a significant impact of AFQ056 on language development in children with fragile X syndrome aged 3-6 years. Objectives The current analyses from biospecimens collected in the FXLEARN study aimed to determine whether AFQ056 affects the level of potential biomarkers associated with Akt/mTOR and matrix metalloproteinase 9 signaling in young individuals with fragile X syndrome. Previous research has indicated that these biomarkers play crucial roles in the pathophysiology of fragile X syndrome. Design A double-blind placebo-controlled parallel-group flexible-dose forced titration design. Methods Blood samples for biomarkers were collected during the FXLEARN at baseline and subsequent visits (1- and 8-month visits). Biomarker analyses included fragile X messenger ribonucleoprotein-1 genotyping by Southern blot and PCR approaches, fragile X messenger ribonucleoprotein-1 mRNA levels determined by PCR, matrix metalloproteinase 9 levels' detection using a magnetic bead panel, and targets of the Akt/mTOR signaling pathway with their phosphorylation levels detected. Results This research revealed that administering AFQ056 does not affect the expression levels of the investigated blood biomarkers in young children with fragile X syndrome. Conclusion Our findings of the lack of association between clinical improvement and biomarkers' levels in the treatment group are in line with the lack of benefit observed in the FXLEARN study. These findings indicate that AFQ056 does not provide benefits as assessed by primary or secondary endpoints. Registration ClincalTrials.gov NCT02920892.
Collapse
Affiliation(s)
- Dragana Protic
- Faculty of Medicine, Department of Pharmacology, Clinical Pharmacology, and Toxicology, University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Elizabeth Breeze
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Sacramento, CA, USA
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Leonard Abbeduto
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | | | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences, Anatomy, and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | | | - Stephanie Morris
- Washington University Medical Center, Saint Louis Children’s Hospital, St. Louis, MO, USA
| | | | - Lisa Prock
- Boston Children’s Hospital, Boston, MA, USA
| | - Amy Talboy
- Emory University Medical Center, Atlanta, GA, USA
| | | | - Jeremy Veenstra-VanderWeele
- Center for Autism and the Developing Brain, New York-Presbyterian, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Flora Tassone
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
22
|
Lakhani A, Huang W, Rodgers CC, Wenner P. Whisker deprivation triggers a distinct form of cortical homeostatic plasticity that is impaired in the Fmr1 KO. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614487. [PMID: 39386532 PMCID: PMC11463509 DOI: 10.1101/2024.09.23.614487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mouse models of Fragile X Syndrome (FXS) have demonstrated impairments in excitatory and inhibitory sensory-evoked neuronal firing. Homeostatic plasticity, which encompasses a set of mechanisms to stabilize baseline activity levels, does not compensate for these changes in activity. Previous work has shown that impairments in homeostatic plasticity are observed in FXS, including deficits in synaptic scaling and intrinsic excitability. Here, we aimed to examine how homeostatic plasticity is altered in vivo in an Fmr1 KO mouse model following unilateral whisker deprivation (WD). We show that WD in the wild type leads to an increase in the proportion of L5/6 somatosensory neurons that are recruited, but this does not occur in the KO. In addition, we observed a change in the threshold of excitatory neurons at a later developmental stage in the KO. Compromised homeostatic plasticity in development could influence sensory processing and long-term cortical organization.
Collapse
|
23
|
Willim J, Woike D, Greene D, Das S, Pfeifer K, Yuan W, Lindsey A, Itani O, Böhme AL, Tibbe D, Hönck HH, Hassani Nia F, Zech M, Brunet T, Faivre L, Sorlin A, Vitobello A, Smol T, Colson C, Baranano K, Schatz K, Bayat A, Schoch K, Spillmann R, Davis EE, Conboy E, Vetrini F, Platzer K, Neuser S, Gburek-Augustat J, Grace AN, Mitchell B, Stegmann A, Sinnema M, Meeks N, Saunders C, Cadieux-Dion M, Hoyer J, Van-Gils J, de Sainte-Agathe JM, Thompson ML, Bebin EM, Weisz-Hubshman M, Tabet AC, Verloes A, Levy J, Latypova X, Harder S, Silverman GA, Pak SC, Schedl T, Freson K, Mumford A, Turro E, Schlein C, Shashi V, Kreienkamp HJ. Variants in LRRC7 lead to intellectual disability, autism, aggression and abnormal eating behaviors. Nat Commun 2024; 15:7909. [PMID: 39256359 PMCID: PMC11387733 DOI: 10.1038/s41467-024-52095-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] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Members of the leucine rich repeat (LRR) and PDZ domain (LAP) protein family are essential for animal development and histogenesis. Densin-180, encoded by LRRC7, is the only LAP protein selectively expressed in neurons. Densin-180 is a postsynaptic scaffold at glutamatergic synapses, linking cytoskeletal elements with signalling proteins such as the α-subunit of Ca2+/calmodulin-dependent protein kinase II. We have previously observed an association between high impact variants in LRRC7 and Intellectual Disability; also three individual cases with variants in LRRC7 had been described. We identify here 33 individuals (one of them previously described) with a dominant neurodevelopmental disorder due to heterozygous missense or loss-of-function variants in LRRC7. The clinical spectrum involves intellectual disability, autism, ADHD, aggression and, in several cases, hyperphagia-associated obesity. A PDZ domain variant interferes with synaptic targeting of Densin-180 in primary cultured neurons. Using in vitro systems (two hybrid, BioID, coimmunoprecipitation of tagged proteins from 293T cells) we identified new candidate interaction partners for the LRR domain, including protein phosphatase 1 (PP1), and observed that variants in the LRR reduced binding to these proteins. We conclude that LRRC7 encodes a major determinant of intellectual development and behaviour.
Collapse
Affiliation(s)
- Jana Willim
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Woike
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Greene
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarada Das
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kevin Pfeifer
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Weimin Yuan
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Anika Lindsey
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Omar Itani
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Amber L Böhme
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Debora Tibbe
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Hinrich Hönck
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fatemeh Hassani Nia
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Zech
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Theresa Brunet
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Laurence Faivre
- Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, CHU Dijon-Bourgogne, Dijon, France
- INSERM-Université de Bourgogne-UMR1231 GAD, Dijon, France
| | - Arthur Sorlin
- INSERM-Université de Bourgogne-UMR1231 GAD, Dijon, France
- Laboratoire de Génomique médicale, Centre NEOMICS, CHU Dijon Bourgogne, Dijon, France
| | - Antonio Vitobello
- INSERM-Université de Bourgogne-UMR1231 GAD, Dijon, France
- Laboratoire de Génomique médicale, Centre NEOMICS, CHU Dijon Bourgogne, Dijon, France
| | - Thomas Smol
- Univ. Lille, CHU Lille, ULR7364 - RADEME, Lille, France
| | - Cindy Colson
- Univ. Lille, CHU Lille, ULR7364 - RADEME, Lille, France
| | - Kristin Baranano
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Krista Schatz
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Allan Bayat
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Center, Dianalund, Denmark
- Department for Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kelly Schoch
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Rebecca Spillmann
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Erica E Davis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Departments of Pediatrics and Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Erin Conboy
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Sonja Neuser
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Janina Gburek-Augustat
- Division of Neuropaediatrics, Hospital for Children and Adolescents, University of Leipzig Medical Center, Leipzig, Germany
| | - Alexandra Noel Grace
- Molecular and Human Genetics Department, Baylor College of Medicine, Houston, TX, USA
| | - Bailey Mitchell
- Baylor College of Medicine in San Antonio, San Antonio, TX, USA
| | - Alexander Stegmann
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Margje Sinnema
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Naomi Meeks
- Children's Hospital Colorado, Division of Clinical Genetics & Metabolism, Aurora, CO, USA
| | - Carol Saunders
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO, USA
- School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Maxime Cadieux-Dion
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO, USA
| | - Juliane Hoyer
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julien Van-Gils
- Genetics Lab, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | | | | | | | - Monika Weisz-Hubshman
- Molecular and Human Genetics Department, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, Tx, USA
| | - Anne-Claude Tabet
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - Alain Verloes
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - Jonathan Levy
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - Xenia Latypova
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - Sönke Harder
- Mass spectrometry and Proteome Analytics, Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gary A Silverman
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Tim Schedl
- Department of Genetics, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Andrew Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Ernest Turro
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian Schlein
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vandana Shashi
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Hans-Jürgen Kreienkamp
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
24
|
Proteau-Lemieux M, Knoth IS, Davoudi S, Martin CO, Bélanger AM, Fontaine V, Côté V, Agbogba K, Vachon K, Whitlock K, Biag HMB, Thurman AJ, Rosenfelt C, Tassone F, Frei J, Capano L, Abbeduto L, Jacquemont S, Hessl D, Hagerman RJ, Schneider A, Bolduc F, Anagnostou E, Lippe S. Specific EEG resting state biomarkers in FXS and ASD. J Neurodev Disord 2024; 16:53. [PMID: 39251926 PMCID: PMC11382468 DOI: 10.1186/s11689-024-09570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) and autism spectrum disorder (ASD) are neurodevelopmental conditions that often have a substantial impact on daily functioning and quality of life. FXS is the most common cause of inherited intellectual disability (ID) and the most common monogenetic cause of ASD. Previous literature has shown that electrophysiological activity measured by electroencephalogram (EEG) during resting state is perturbated in FXS and ASD. However, whether electrophysiological profiles of participants with FXS and ASD are similar remains unclear. The aim of this study was to compare EEG alterations found in these two clinical populations presenting varying degrees of cognitive and behavioral impairments. METHODS Resting state EEG signal complexity, alpha peak frequency (APF) and power spectral density (PSD) were compared between 47 participants with FXS (aged between 5-20), 49 participants with ASD (aged between 6-17), and 52 neurotypical (NT) controls with a similar age distribution using MANCOVAs with age as covariate when appropriate. MANCOVAs controlling for age, when appropriate, and nonverbal intelligence quotient (NVIQ) score were subsequently performed to determine the impact of cognitive functioning on EEG alterations. RESULTS Our results showed that FXS participants manifested decreased signal complexity and APF compared to ASD participants and NT controls, as well as altered power in the theta, alpha and low gamma frequency bands. ASD participants showed exaggerated beta power compared to FXS participants and NT controls, as well as enhanced low and high gamma power compared to NT controls. However, ASD participants did not manifest altered signal complexity or APF. Furthermore, when controlling for NVIQ, results of decreased complexity in higher scales and lower APF in FXS participants compared to NT controls and ASD participants were not replicated. CONCLUSIONS These findings suggest that signal complexity and APF might reflect cognitive functioning, while altered power in the low gamma frequency band might be associated with neurodevelopmental conditions, particularly FXS and ASD.
Collapse
Affiliation(s)
- Mélodie Proteau-Lemieux
- Department of Psychology, University of Montreal, Montreal, QC, Canada
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Inga Sophia Knoth
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Saeideh Davoudi
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | | | - Anne-Marie Bélanger
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Valérie Fontaine
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Valérie Côté
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Kristian Agbogba
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | | | | | - Hazel Maridith Barlahan Biag
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Angela John Thurman
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Cory Rosenfelt
- Department of Pediatric Neurology, University of Alberta, Edmonton, AB, Canada
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Julia Frei
- McMaster University of Ottawa, Ottawa, ON, Canada
| | - Lucia Capano
- Queen's University of Kingston, Kingston, ON, Canada
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Sébastien Jacquemont
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - David Hessl
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Randi Jenssen Hagerman
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Andrea Schneider
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Francois Bolduc
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Evdokia Anagnostou
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
- Holland Bloorview Research Center, Toronto, ON, Canada
| | - Sarah Lippe
- Department of Psychology, University of Montreal, Montreal, QC, Canada.
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada.
| |
Collapse
|
25
|
Attallah A, Ardourel M, Gallazzini F, Lesne F, De Oliveira A, Togbé D, Briault S, Perche O. Lack of FMRP in the retina: Evidence of a retinal specific transcriptomic profile. Exp Eye Res 2024; 246:110015. [PMID: 39089568 DOI: 10.1016/j.exer.2024.110015] [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: 03/27/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Fragile X Syndrome (FXS), the most common inherited form of human intellectual disability, is a monogenic neurodevelopmental disorder caused by a loss-of-function mutation of the FMR1 gene. FMR1 is encoding the Fragile X Messenger Ribonucleo Protein (FMRP) an RNA-binding protein that regulates the translation of synaptic proteins. The absence of FMRP expression has many important consequences on synaptic plasticity and function, leading to the FXS clinical phenotype. Over the last decade, a visual neurosensorial phenotype had been described in the FXS patients as well as in the murine model (Fmr1-/ymice), characterized by retinal deficits associated to retinal perception alterations. However, although the transcriptomic profile in the absence of FMRP has been studied in the cerebral part of the central nervous system (CNS), there are no actual data for the retina which is an extension of the CNS. Herein, we investigate the transcriptomic profile of mRNA from whole retinas of Fmr1-/ymice. Interestingly, we found a specific signature of Fmrp absence on retinal mRNA expression with few common genes compared to other brain studies. Gene Ontology on these retinal specific genes demonstrated an enrichment in retinal development genes as well as in synaptic genes. These alterations could be linked to the reported retinal phenotype of the FXS condition. In conclusion, we describe for the first time, retinal-specific transcriptomic changes in the absence of FMRP.
Collapse
Affiliation(s)
- Amir Attallah
- Orléans University, University Hospital Center of Orleans, LI(2)RSO, 14, Avenue de l'hôpital, 45100, Orléans, France; Orleans University, CNRS, laboratoire INEM, UMR7355, 3b Rue de la Férollerie, F-45071, Orléans, Cedex 2, France; ART ARNm US55, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Maryvonne Ardourel
- Orléans University, University Hospital Center of Orleans, LI(2)RSO, 14, Avenue de l'hôpital, 45100, Orléans, France; ART ARNm US55, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Felix Gallazzini
- University Hospital Center of Orleans, Genetic Department, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Fabien Lesne
- University Hospital Center of Orléans CAR&IB, Pôle Biopatholgie, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Anthony De Oliveira
- University Hospital Center of Orléans CAR&IB, Pôle Biopatholgie, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Dieudonnée Togbé
- Orleans University, CNRS, laboratoire INEM, UMR7355, 3b Rue de la Férollerie, F-45071, Orléans, Cedex 2, France
| | - Sylvain Briault
- Orléans University, University Hospital Center of Orleans, LI(2)RSO, 14, Avenue de l'hôpital, 45100, Orléans, France; ART ARNm US55, 14 Avenue de l'Hôpital, 45100, Orléans, France; University Hospital Center of Orleans, Genetic Department, 14 Avenue de l'Hôpital, 45100, Orléans, France
| | - Olivier Perche
- Orléans University, University Hospital Center of Orleans, LI(2)RSO, 14, Avenue de l'hôpital, 45100, Orléans, France; ART ARNm US55, 14 Avenue de l'Hôpital, 45100, Orléans, France; University Hospital Center of Orleans, Genetic Department, 14 Avenue de l'Hôpital, 45100, Orléans, France.
| |
Collapse
|
26
|
Morin-Parent F, Champigny C, Côté S, Mohamad T, Hasani SA, Çaku A, Corbin F, Lepage JF. Neurophysiological effects of a combined treatment of lovastatin and minocycline in patients with fragile X syndrome: Ancillary results of the LOVAMIX randomized clinical trial. Autism Res 2024; 17:1944-1956. [PMID: 39248107 DOI: 10.1002/aur.3222] [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: 06/04/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Fragile X syndrome (FXS) is the primary hereditary cause of intellectual disability and autism spectrum disorder. It is characterized by exacerbated neuronal excitability, and its correction is considered an objective measure of treatment response in animal models, a marker albeit rarely used in clinical trials. Here, we used an extensive transcranial magnetic stimulation (TMS) battery to assess the neurophysiological effects of a therapy combining two disease-modifying drugs, lovastatin (40 mg) and minocycline (100 mg), administered alone for 8 weeks and in combination for 12 weeks, in 19 patients (mean age of 23.58 ± 1.51) with FXS taking part in the LOVAmix trial. The TMS battery, which included the resting motor threshold, short-interval intracortical inhibition, long-interval intracortical inhibition, corticospinal silent period, and intracortical facilitation, was completed at baseline after 8 weeks of monotherapy (visit 2 of the clinical trial) and after 12 weeks of dual therapy (visit 4 of the clinical trial). Repeated measure ANOVAs were performed between baseline and visit 2 (monotherapy) and visit 3 (dual therapy) with interactions for which monotherapy the participants received when they began the clinical trial. Results showed that dual therapy was associated with reduced cortical excitability after 20 weeks. This was reflected by a significant increase in the resting-motor threshold after dual therapy compared to baseline. There was a tendency for enhanced short-intracortical inhibition, a marker of GABAa-mediated inhibition after 8 weeks of monotherapy compared to baseline. Together, these results suggest that a combined therapy of minocycline and lovastatin might act on the core neurophysiopathology of FXS. This trial was registered at clinicaltrials.gov (NCT02680379).
Collapse
Affiliation(s)
- Florence Morin-Parent
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Camille Champigny
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - Samantha Côté
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Teddy Mohamad
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Seyede Anis Hasani
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Artuela Çaku
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - François Corbin
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - Jean-François Lepage
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| |
Collapse
|
27
|
Liu W, Chen QY, Li XH, Zhou Z, Zhuo M. Cortical Tagged Synaptic Long-Term Depression in the Anterior Cingulate Cortex of Adult Mice. J Neurosci 2024; 44:e0028242024. [PMID: 39054067 PMCID: PMC11358531 DOI: 10.1523/jneurosci.0028-24.2024] [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: 01/05/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
The anterior cingulate cortex (ACC) is a key cortical region for pain perception and emotion. Different forms of synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), have been reported in the ACC. Synaptic tagging of LTP plays an important role in hippocampus-related associative memory. In this study, we demonstrate that synaptic tagging of LTD is detected in the ACC of adult male and female mice. This form of tagged LTD requires the activation of metabotropic glutamate receptor subtype 1 (mGluR1). The induction of tagged LTD is time-related with the strongest tagged LTD appearing when the interval between two independent stimuli is 30 min. Inhibitors of mGluR1 blocked the induction of tagged LTD; however, blocking N-methyl-d-aspartate receptors did not affect the induction of tagged LTD. Nimodipine, an inhibitor of L-type voltage-gated calcium channels, also blocked tagged LTD. In an animal model of amputation, we found that tagged LTD was either reduced or completely blocked. Together with our previous report of tagged LTP in the ACC, this study strongly suggests that excitatory synapses in the adult ACC are highly plastic. The biphasic tagging of synaptic transmission provides a new form of heterosynaptic plasticity in the ACC which has functional and pathophysiological significance in phantom pain.
Collapse
Affiliation(s)
- Weiqi Liu
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Zhuomin Institute of Brain Research, Qingdao International Academician Park, Qingdao 266000, China
| | - Qi-Yu Chen
- Zhuomin Institute of Brain Research, Qingdao International Academician Park, Qingdao 266000, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xu-Hui Li
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Zhuomin Institute of Brain Research, Qingdao International Academician Park, Qingdao 266000, China
| | - Zhaoxiang Zhou
- Zhuomin Institute of Brain Research, Qingdao International Academician Park, Qingdao 266000, China
- Department of Exercise & Health Science, Xi'an Physical Education University, Xi'an 710068, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Zhuomin Institute of Brain Research, Qingdao International Academician Park, Qingdao 266000, China
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
28
|
Rani R, Sri NS, Medishetti R, Chatti K, Sevilimedu A. Loss of FMRP affects ovarian development and behaviour through multiple pathways in a zebrafish model of fragile X syndrome. Hum Mol Genet 2024; 33:1391-1405. [PMID: 38710511 PMCID: PMC7616351 DOI: 10.1093/hmg/ddae077] [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: 02/16/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
Fragile X syndrome (FXS) is an inherited neurodevelopmental disorder and the leading genetic cause of autism spectrum disorders. FXS is caused by loss of function mutations in Fragile X mental retardation protein (FMRP), an RNA binding protein that is known to regulate translation of its target mRNAs, predominantly in the brain and gonads. The molecular mechanisms connecting FMRP function to neurodevelopmental phenotypes are well understood. However, neither the full extent of reproductive phenotypes, nor the underlying molecular mechanisms have been as yet determined. Here, we developed new fmr1 knockout zebrafish lines and show that they mimic key aspects of FXS neuronal phenotypes across both larval and adult stages. Results from the fmr1 knockout females also showed that altered gene expression in the brain, via the neuroendocrine pathway contribute to distinct abnormal phenotypes during ovarian development and oocyte maturation. We identified at least three mechanisms underpinning these defects, including altered neuroendocrine signaling in sexually mature females resulting in accelerated ovarian development, altered expression of germ cell and meiosis promoting genes at various stages during oocyte maturation, and finally a strong mitochondrial impairment in late stage oocytes from knockout females. Our findings have implications beyond FXS in the study of reproductive function and female infertility. Dissection of the translation control pathways during ovarian development using models like the knockout lines reported here may reveal novel approaches and targets for fertility treatments.
Collapse
Affiliation(s)
- Rita Rani
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
| | - N Sushma Sri
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
| | - Raghavender Medishetti
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
| | - Kiranam Chatti
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
- Center for Rare Disease Models, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
| | - Aarti Sevilimedu
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
- Center for Rare Disease Models, Dr. Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana500046, India
| |
Collapse
|
29
|
Han Y, Jia Z, Xu K, Li Y, Lu S, Guan L. CRISPR-Cpf1 system and its applications in animal genome editing. Mol Genet Genomics 2024; 299:75. [PMID: 39085660 DOI: 10.1007/s00438-024-02166-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] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR) and their associated protein (Cas) system is a gene editing technology guided by RNA endonuclease. The CRISPR-Cas12a (also known as CRISPR-Cpf1) system is extensively utilized in genome editing research due to its accuracy and high efficiency. In this paper, we primarily focus on the application of CRISPR-Cpf1 technology in the construction of disease models and gene therapy. Firstly, the structure and mechanism of the CRISPR-Cas system are introduced. Secondly, the similarities and differences between CRISPR-Cpf1 and CRISPR-Cas9 technologies are compared. Thirdly, the main focus is on the application of the CRISPR-Cpf1 system in cell and animal genome editing. Finally, the challenges faced by CRISPR-Cpf1 technology and corresponding strategies are analyzed. Although CRISPR-Cpf1 technology has certain off-target effects, it can effectively and accurately edit cell and animal genomes, and has significant advantages in the preclinical research.
Collapse
Affiliation(s)
- Yawei Han
- College of Tobacco Science and Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, Henan, China
| | - Zisen Jia
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Number 601, Jinsui Road, Xinxiang, 453003, Henan, China
| | - Keli Xu
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Number 601, Jinsui Road, Xinxiang, 453003, Henan, China
| | - Yangyang Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Number 601, Jinsui Road, Xinxiang, 453003, Henan, China
| | - Suxiang Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
| | - Lihong Guan
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Number 601, Jinsui Road, Xinxiang, 453003, Henan, China.
| |
Collapse
|
30
|
Dionne O, Abolghasemi A, Corbin F, Çaku A. Implication of the endocannabidiome and metabolic pathways in fragile X syndrome pathophysiology. Psychiatry Res 2024; 337:115962. [PMID: 38763080 DOI: 10.1016/j.psychres.2024.115962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
Fragile X Syndrome (FXS) results from the silencing of the FMR1 gene and is the most prevalent inherited cause of intellectual disability and the most frequent monogenic cause of autism spectrum disorder. It is well established that Fragile X individuals are subjected to a wide array of comorbidities, ranging from cognitive, behavioural, and medical origin. Furthermore, recent studies have also described metabolic impairments in FXS individuals. However, the molecular mechanisms linking FMRP deficiency to improper metabolism are still misunderstood. The endocannabinoidome (eCBome) is a lipid-based signalling system that regulates several functions across the body, ranging from cognition, behaviour and metabolism. Alterations in the eCBome have been described in FXS animal models and linked to neuronal hyperexcitability, a core deficit of the disease. However, the potential link between dysregulation of the eCBome and altered metabolism observed in FXS remains unexplored. As such, this review aims to overcome this issue by describing the most recent finding related to eCBome and metabolic dysfunctions in the context of FXS. A better comprehension of this association will help deepen our understanding of FXS pathophysiology and pave the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Olivier Dionne
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada.
| | - Armita Abolghasemi
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| | - François Corbin
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| | - Artuela Çaku
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Canada
| |
Collapse
|
31
|
Dwivedi D, Dumontier D, Sherer M, Lin S, Mirow AMC, Qiu Y, Xu Q, Liebman SA, Joseph D, Datta SR, Fishell G, Pouchelon G. Metabotropic signaling within somatostatin interneurons controls transient thalamocortical inputs during development. Nat Commun 2024; 15:5421. [PMID: 38926335 PMCID: PMC11208423 DOI: 10.1038/s41467-024-49732-w] [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: 12/18/2023] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
During brain development, neural circuits undergo major activity-dependent restructuring. Circuit wiring mainly occurs through synaptic strengthening following the Hebbian "fire together, wire together" precept. However, select connections, essential for circuit development, are transient. They are effectively connected early in development, but strongly diminish during maturation. The mechanisms by which transient connectivity recedes are unknown. To investigate this process, we characterize transient thalamocortical inputs, which depress onto somatostatin inhibitory interneurons during development, by employing optogenetics, chemogenetics, transcriptomics and CRISPR-based strategies in mice. We demonstrate that in contrast to typical activity-dependent mechanisms, transient thalamocortical connectivity onto somatostatin interneurons is non-canonical and involves metabotropic signaling. Specifically, metabotropic-mediated transcription, of guidance molecules in particular, supports the elimination of this connectivity. Remarkably, we found that this process impacts the development of normal exploratory behaviors of adult mice.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA
| | | | - Mia Sherer
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA
| | - Sherry Lin
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
| | - Andrea M C Mirow
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Harbor, NY, USA
| | - Yanjie Qiu
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA
| | - Qing Xu
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA
- Center for Genomics & Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Samuel A Liebman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Harbor, NY, USA
| | - Djeckby Joseph
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Harbor, NY, USA
| | - Sandeep R Datta
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA
| | - Gord Fishell
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA.
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA.
| | - Gabrielle Pouchelon
- Harvard Medical School, Department of Neurobiology, Boston, MA, USA.
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA, USA.
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Harbor, NY, USA.
| |
Collapse
|
32
|
Gholipour P, Ebrahimi Z, Mohammadkhani R, Ghahremani R, Salehi I, Sarihi A, Komaki A, Karimi SA. Effects of (S)-3,4-DCPG, an mGlu8 receptor agonist, on hippocampal long-term potentiation at perforant pathway-dentate gyrus synapses in prenatal valproic acid-induced rat model of autism. Sci Rep 2024; 14:13168. [PMID: 38849397 PMCID: PMC11161498 DOI: 10.1038/s41598-024-63728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
Autism spectrum disorder (ASD) is a pervasive neurodevelopmental condition characterized by social interaction deficits, communication impairments, repetitive behaviors, and sensory sensitivities. While the etiology of ASD is multifaceted, abnormalities in glutamatergic neurotransmission and synaptic plasticity have been implicated. This study investigated the role of metabotropic glutamate receptor 8 (mGlu8) in modulating long-term potentiation (LTP) in a rat model of ASD induced by prenatal valproic acid (VPA) exposure. To induce an animal model with autism-like characteristics, pregnant rats received an intraperitoneal injection of 500 mg/kg of sodium valproate (NaVPA) on embryonic day 12.5. High-frequency stimulation was applied to the perforant path-dentate gyrus (PP-DG) synapse to induce LTP, while the mGlu8 receptor agonist (S)-3,4-dicarboxyphenylglycine (DCPG) was administered into the DG. The results revealed that VPA-exposed rats exhibited reduced LTP compared to controls. DCPG had contrasting effects, inhibiting LTP in controls and enhancing it in VPA-exposed rats. Moreover, reduced social novelty preference index (SNPI) in VPA-exposed rats was reversed by intra-DG administration of S-3,4-DCPG. In conclusion, our study advances our understanding of the complex relationship between glutamatergic neurotransmission, synaptic plasticity, and VPA-induced autism model. The findings suggest that mGlu8 receptor dysfunction plays a role in the impaired synaptic plasticity seen in ASD.
Collapse
Affiliation(s)
- Parsa Gholipour
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Zahra Ebrahimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Reihaneh Mohammadkhani
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Reza Ghahremani
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Birjand, Birjand, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
33
|
Coulson RL, Mourrain P, Wang GX. The intersection of sleep and synaptic translation in synaptic plasticity deficits in neurodevelopmental disorders. J Comp Physiol B 2024; 194:253-263. [PMID: 38396062 PMCID: PMC11233386 DOI: 10.1007/s00360-023-01531-3] [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: 08/28/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 02/25/2024]
Abstract
Individuals with neurodevelopmental disorders experience persistent sleep deficits, and there is increasing evidence that sleep dysregulation is an underlying cause, rather than merely an effect, of the synaptic and behavioral defects observed in these disorders. At the molecular level, dysregulation of the synaptic proteome is a common feature of neurodevelopmental disorders, though the mechanism connecting these molecular and behavioral phenotypes is an ongoing area of investigation. A role for eIF2α in shifting the local proteome in response to changes in the conditions at the synapse has emerged. Here, we discuss recent progress in characterizing the intersection of local synaptic translation and sleep and propose a reciprocal mechanism of dysregulation in the development of synaptic plasticity defects in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rochelle L Coulson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- INSERM 1024, Ecole Normale Supérieure, Paris, France
| | - Gordon X Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Tran H, Le L, Singh BN, Kramer J, Steward R. Tet controls axon guidance in early brain development through glutamatergic signaling. iScience 2024; 27:109634. [PMID: 38655199 PMCID: PMC11035372 DOI: 10.1016/j.isci.2024.109634] [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: 08/14/2023] [Revised: 12/18/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in ten-eleven translocation (TET) proteins are associated with human neurodevelopmental disorders. We find a function of Tet in regulating Drosophila early brain development. The Tet DNA-binding domain (TetAXXC) is required for axon guidance in the mushroom body (MB). Glutamine synthetase 2 (Gs2), a key enzyme in glutamatergic signaling, is significantly down-regulated in the TetAXXC brains. Loss of Gs2 recapitulates the TetAXXC phenotype. Surprisingly, Tet and Gs2 act in the insulin-producing cells (IPCs) to control MB axon guidance, and overexpression of Gs2 in IPCs rescues the defects of TetAXXC. Feeding TetAXXC with metabotropic glutamate receptor antagonist MPEP rescues the phenotype while glutamate enhances it. Mutants in Tet and Drosophila Fmr1, the homolog of human FMR1, have similar defects, and overexpression of Gs2 in IPCs also rescues the Fmr1 phenotype. We provide the first evidence that Tet controls the guidance of developing brain axons by modulating glutamatergic signaling.
Collapse
Affiliation(s)
- Hiep Tran
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Le Le
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Badri Nath Singh
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Joseph Kramer
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Ruth Steward
- Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
- Department of Molecular Biology and Biochemistry, Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
35
|
Croom K, Rumschlag JA, Erickson MA, Binder D, Razak KA. Sex differences during development in cortical temporal processing and event related potentials in wild-type and fragile X syndrome model mice. J Neurodev Disord 2024; 16:24. [PMID: 38720271 PMCID: PMC11077726 DOI: 10.1186/s11689-024-09539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is currently diagnosed in approximately 1 in 44 children in the United States, based on a wide array of symptoms, including sensory dysfunction and abnormal language development. Boys are diagnosed ~ 3.8 times more frequently than girls. Auditory temporal processing is crucial for speech recognition and language development. Abnormal development of temporal processing may account for ASD language impairments. Sex differences in the development of temporal processing may underlie the differences in language outcomes in male and female children with ASD. To understand mechanisms of potential sex differences in temporal processing requires a preclinical model. However, there are no studies that have addressed sex differences in temporal processing across development in any animal model of ASD. METHODS To fill this major gap, we compared the development of auditory temporal processing in male and female wildtype (WT) and Fmr1 knock-out (KO) mice, a model of Fragile X Syndrome (FXS), a leading genetic cause of ASD-associated behaviors. Using epidural screw electrodes, we recorded auditory event related potentials (ERP) and auditory temporal processing with a gap-in-noise auditory steady state response (ASSR) paradigm at young (postnatal (p)21 and p30) and adult (p60) ages from both auditory and frontal cortices of awake, freely moving mice. RESULTS The results show that ERP amplitudes were enhanced in both sexes of Fmr1 KO mice across development compared to WT counterparts, with greater enhancement in adult female than adult male KO mice. Gap-ASSR deficits were seen in the frontal, but not auditory, cortex in early development (p21) in female KO mice. Unlike male KO mice, female KO mice show WT-like temporal processing at p30. There were no temporal processing deficits in the adult mice of both sexes. CONCLUSIONS These results show a sex difference in the developmental trajectories of temporal processing and hypersensitive responses in Fmr1 KO mice. Male KO mice show slower maturation of temporal processing than females. Female KO mice show stronger hypersensitive responses than males later in development. The differences in maturation rates of temporal processing and hypersensitive responses during various critical periods of development may lead to sex differences in language function, arousal and anxiety in FXS.
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
| | - Michael A Erickson
- Department of Psychology, University of California, 900 University Avenue, Riverside, USA
| | - Devin 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, 900 University Avenue, Riverside, USA.
| |
Collapse
|
36
|
Stamenkovic V, Lautz JD, Harsh FM, Smith SEP. SRC family kinase inhibition rescues molecular and behavioral phenotypes, but not protein interaction network dynamics, in a mouse model of Fragile X syndrome. Mol Psychiatry 2024; 29:1392-1405. [PMID: 38297084 PMCID: PMC11524049 DOI: 10.1038/s41380-024-02418-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024]
Abstract
Glutamatergic synapses encode information from extracellular inputs using dynamic protein interaction networks (PINs) that undergo widespread reorganization following synaptic activity, allowing cells to distinguish between signaling inputs and generate coordinated cellular responses. Here, we investigate how Fragile X Messenger Ribonucleoprotein (FMRP) deficiency disrupts signal transduction through a glutamatergic synapse PIN downstream of NMDA receptor or metabotropic glutamate receptor (mGluR) stimulation. In cultured cortical neurons or acute cortical slices from P7, P17 and P60 FMR1-/y mice, the unstimulated protein interaction network state resembled that of wildtype littermates stimulated with mGluR agonists, demonstrating resting state pre-activation of mGluR signaling networks. In contrast, interactions downstream of NMDAR stimulation were similar to WT. We identified the Src family kinase (SFK) Fyn as a network hub, because many interactions involving Fyn were pre-activated in FMR1-/y animals. We tested whether targeting SFKs in FMR1-/y mice could modify disease phenotypes, and found that Saracatinib (SCB), an SFK inhibitor, normalized elevated basal protein synthesis, novel object recognition memory and social behavior in FMR1-/y mice. However, SCB treatment did not normalize the PIN to a wild-type-like state in vitro or in vivo, but rather induced extensive changes to protein complexes containing Shank3, NMDARs and Fyn. We conclude that targeting abnormal nodes of a PIN can identify potential disease-modifying drugs, but behavioral rescue does not correlate with PIN normalization.
Collapse
Affiliation(s)
- Vera Stamenkovic
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Felicia M Harsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| |
Collapse
|
37
|
Lee S, Kim J, Ryu HH, Jang H, Lee D, Lee S, Song JM, Lee YS, Ho Suh Y. SHP2 regulates GluA2 tyrosine phosphorylation required for AMPA receptor endocytosis and mGluR-LTD. Proc Natl Acad Sci U S A 2024; 121:e2316819121. [PMID: 38657042 PMCID: PMC11066993 DOI: 10.1073/pnas.2316819121] [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: 10/02/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Posttranslational modifications regulate the properties and abundance of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors that mediate fast excitatory synaptic transmission and synaptic plasticity in the central nervous system. During long-term depression (LTD), protein tyrosine phosphatases (PTPs) dephosphorylate tyrosine residues in the C-terminal tail of AMPA receptor GluA2 subunit, which is essential for GluA2 endocytosis and group I metabotropic glutamate receptor (mGluR)-dependent LTD. However, as a selective downstream effector of mGluRs, the mGluR-dependent PTP responsible for GluA2 tyrosine dephosphorylation remains elusive at Schaffer collateral (SC)-CA1 synapses. In the present study, we find that mGluR5 stimulation activates Src homology 2 (SH2) domain-containing phosphatase 2 (SHP2) by increasing phospho-Y542 levels in SHP2. Under steady-state conditions, SHP2 plays a protective role in stabilizing phospho-Y869 of GluA2 by directly interacting with GluA2 phosphorylated at Y869, without affecting GluA2 phospho-Y876 levels. Upon mGluR5 stimulation, SHP2 dephosphorylates GluA2 at Y869 and Y876, resulting in GluA2 endocytosis and mGluR-LTD. Our results establish SHP2 as a downstream effector of mGluR5 and indicate a dual action of SHP2 in regulating GluA2 tyrosine phosphorylation and function. Given the implications of mGluR5 and SHP2 in synaptic pathophysiology, we propose SHP2 as a promising therapeutic target for neurodevelopmental and autism spectrum disorders.
Collapse
Affiliation(s)
- Sanghyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jungho Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hyun-Hee Ryu
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hanbyul Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - DoEun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Seungha Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jae-man Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Yong-Seok Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| |
Collapse
|
38
|
Dwivedi D, Dumontier D, Sherer M, Lin S, Mirow AM, Qiu Y, Xu Q, Liebman SA, Joseph D, Datta SR, Fishell G, Pouchelon G. Metabotropic signaling within somatostatin interneurons controls transient thalamocortical inputs during development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.21.558862. [PMID: 37790336 PMCID: PMC10542166 DOI: 10.1101/2023.09.21.558862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
During brain development, neural circuits undergo major activity-dependent restructuring. Circuit wiring mainly occurs through synaptic strengthening following the Hebbian "fire together, wire together" precept. However, select connections, essential for circuit development, are transient. They are effectively connected early in development, but strongly diminish during maturation. The mechanisms by which transient connectivity recedes are unknown. To investigate this process, we characterize transient thalamocortical inputs, which depress onto somatostatin inhibitory interneurons during development, by employing optogenetics, chemogenetics, transcriptomics and CRISPR-based strategies. We demonstrate that in contrast to typical activity-dependent mechanisms, transient thalamocortical connectivity onto somatostatin interneurons is non-canonical and involves metabotropic signaling. Specifically, metabotropic-mediated transcription, of guidance molecules in particular, supports the elimination of this connectivity. Remarkably, we found that this developmental process impacts the development of normal exploratory behaviors of adult mice.
Collapse
|
39
|
Meneghetti N, Vannini E, Mazzoni A. Rodents' visual gamma as a biomarker of pathological neural conditions. J Physiol 2024; 602:1017-1048. [PMID: 38372352 DOI: 10.1113/jp283858] [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: 12/13/2022] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
Neural gamma oscillations (indicatively 30-100 Hz) are ubiquitous: they are associated with a broad range of functions in multiple cortical areas and across many animal species. Experimental and computational works established gamma rhythms as a global emergent property of neuronal networks generated by the balanced and coordinated interaction of excitation and inhibition. Coherently, gamma activity is strongly influenced by the alterations of synaptic dynamics which are often associated with pathological neural dysfunctions. We argue therefore that these oscillations are an optimal biomarker for probing the mechanism of cortical dysfunctions. Gamma oscillations are also highly sensitive to external stimuli in sensory cortices, especially the primary visual cortex (V1), where the stimulus dependence of gamma oscillations has been thoroughly investigated. Gamma manipulation by visual stimuli tuning is particularly easy in rodents, which have become a standard animal model for investigating the effects of network alterations on gamma oscillations. Overall, gamma in the rodents' visual cortex offers an accessible probe on dysfunctional information processing in pathological conditions. Beyond vision-related dysfunctions, alterations of gamma oscillations in rodents were indeed also reported in neural deficits such as migraine, epilepsy and neurodegenerative or neuropsychiatric conditions such as Alzheimer's, schizophrenia and autism spectrum disorders. Altogether, the connections between visual cortical gamma activity and physio-pathological conditions in rodent models underscore the potential of gamma oscillations as markers of neuronal (dys)functioning.
Collapse
Affiliation(s)
- Nicolò Meneghetti
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Alberto Mazzoni
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
40
|
Fishburn JLA, Larson HL, Nguyen A, Welch CJ, Moore T, Penn A, Newman J, Mangino A, Widman E, Ghobashy R, Witherspoon J, Lee W, Mulligan KA. Bisphenol F affects neurodevelopmental gene expression, mushroom body development, and behavior in Drosophila melanogaster. Neurotoxicol Teratol 2024; 102:107331. [PMID: 38301979 DOI: 10.1016/j.ntt.2024.107331] [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: 10/03/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/03/2024]
Abstract
Bisphenol F (BPF) is a potential neurotoxicant used as a replacement for bisphenol A (BPA) in polycarbonate plastics and epoxy resins. We investigated the neurodevelopmental impacts of BPF exposure using Drosophila melanogaster as a model. Our transcriptomic analysis indicated that developmental exposure to BPF caused the downregulation of neurodevelopmentally relevant genes, including those associated with synapse formation and neuronal projection. To investigate the functional outcome of BPF exposure, we evaluated neurodevelopmental impacts across two genetic strains of Drosophila- w1118 (control) and the Fragile X Syndrome (FXS) model-by examining both behavioral and neuronal phenotypes. We found that BPF exposure in w1118 Drosophila caused hypoactive larval locomotor activity, decreased time spent grooming by adults, reduced courtship activity, and increased the severity but not frequency of β-lobe midline crossing defects by axons in the mushroom body. In contrast, although BPF reduced peristaltic contractions in FXS larvae, it had no impact on other larval locomotor phenotypes, grooming activity, or courtship activity. Strikingly, BPF exposure reduced both the severity and frequency of β-lobe midline crossing defects in the mushroom body of FXS flies, a phenotype previously observed in FXS flies exposed to BPA. This data indicates that BPF can affect neurodevelopment and its impacts vary depending on genetic background. Further, BPF may elicit a gene-environment interaction with Drosophila fragile X messenger ribonucleoprotein 1 (dFmr1)-the ortholog of human FMR1, which causes fragile X syndrome and is the most common monogenetic cause of intellectual disability and autism spectrum disorder.
Collapse
Affiliation(s)
- Judith L A Fishburn
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Heather L Larson
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - An Nguyen
- Department of Computer Science, College of Natural Sciences and Mathematics, San José State University, 6000 J Street, San José, CA 95819, United States
| | - Chloe J Welch
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Taylor Moore
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Aliyah Penn
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Johnathan Newman
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Anthony Mangino
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Erin Widman
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Rana Ghobashy
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Jocelyn Witherspoon
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States
| | - Wendy Lee
- Department of Computer Science, College of Natural Sciences and Mathematics, San José State University, 6000 J Street, San José, CA 95819, United States
| | - Kimberly A Mulligan
- Department of Biological Sciences, College of Natural Sciences and Mathematics, California State University, 6000 J Street, Sacramento, CA 95819, United States.
| |
Collapse
|
41
|
Chato-Astrain I, Pronot M, Coppola T, Martin S. Molecular Organization and Regulation of the Mammalian Synapse by the Post-Translational Modification SUMOylation. Cells 2024; 13:420. [PMID: 38474384 PMCID: PMC10930594 DOI: 10.3390/cells13050420] [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: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Neurotransmission occurs within highly specialized compartments forming the active synapse where the complex organization and dynamics of the interactions are tightly orchestrated both in time and space. Post-translational modifications (PTMs) are central to these spatiotemporal regulations to ensure an efficient synaptic transmission. SUMOylation is a dynamic PTM that modulates the interactions between proteins and consequently regulates the conformation, the distribution and the trafficking of the SUMO-target proteins. SUMOylation plays a crucial role in synapse formation and stabilization, as well as in the regulation of synaptic transmission and plasticity. In this review, we summarize the molecular consequences of this protein modification in the structural organization and function of the mammalian synapse. We also outline novel activity-dependent regulation and consequences of the SUMO process and explore how this protein modification can functionally participate in the compartmentalization of both pre- and post-synaptic sites.
Collapse
Affiliation(s)
- Isabel Chato-Astrain
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| | - Marie Pronot
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK;
| | - Thierry Coppola
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| | - Stéphane Martin
- Université Côte d’Azur, CNRS, Inserm, IPMC, Sophia Antipolis, F-06560 Valbonne, France; (I.C.-A.); (T.C.)
| |
Collapse
|
42
|
Fang M, Deibler SK, Krishnamurthy PM, Wang F, Rodriguez P, Banday S, Virbasius CM, Sena-Esteves M, Watts JK, Green MR. EZH2 inhibition reactivates epigenetically silenced FMR1 and normalizes molecular and electrophysiological abnormalities in fragile X syndrome neurons. Front Neurosci 2024; 18:1348478. [PMID: 38449737 PMCID: PMC10915284 DOI: 10.3389/fnins.2024.1348478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
Fragile X Syndrome (FXS) is a neurological disorder caused by epigenetic silencing of the FMR1 gene. Reactivation of FMR1 is a potential therapeutic approach for FXS that would correct the root cause of the disease. Here, using a candidate-based shRNA screen, we identify nine epigenetic repressors that promote silencing of FMR1 in FXS cells (called FMR1 Silencing Factors, or FMR1- SFs). Inhibition of FMR1-SFs with shRNAs or small molecules reactivates FMR1 in cultured undifferentiated induced pluripotent stem cells, neural progenitor cells (NPCs) and post-mitotic neurons derived from FXS patients. One of the FMR1-SFs is the histone methyltransferase EZH2, for which an FDA-approved small molecule inhibitor, EPZ6438 (also known as tazemetostat), is available. We show that EPZ6438 substantially corrects the characteristic molecular and electrophysiological abnormalities of cultured FXS neurons. Unfortunately, EZH2 inhibitors do not efficiently cross the blood-brain barrier, limiting their therapeutic use for FXS. Recently, antisense oligonucleotide (ASO)-based approaches have been developed as effective treatment options for certain central nervous system disorders. We therefore derived efficacious ASOs targeting EZH2 and demonstrate that they reactivate FMR1 expression and correct molecular and electrophysiological abnormalities in cultured FXS neurons, and reactivate FMR1 expression in human FXS NPCs engrafted within the brains of mice. Collectively, our results establish EZH2 inhibition in general, and EZH2 ASOs in particular, as a therapeutic approach for FXS.
Collapse
Affiliation(s)
- Minggang Fang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Sara K. Deibler
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | | | - Feng Wang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Paola Rodriguez
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Ching-Man Virbasius
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Michael R. Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
43
|
Malyshev AV, Pavshintcev VV, Mitkin NA, Sukhanova IA, Gedzun VR, Zlobin AS, Doronin II, Babkin GA, Sawyer TK. The novel peptide LCGM-10 attenuates metabotropic glutamate receptor 5 activity and demonstrates behavioral effects in animal models. Front Behav Neurosci 2024; 18:1333258. [PMID: 38385004 PMCID: PMC10879279 DOI: 10.3389/fnbeh.2024.1333258] [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: 11/04/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
We employed a structural bioinformatics approach to develop novel peptides with predicted affinity to the binding site for negative allosteric modulators (NAMs) of metabotropic glutamate receptor 5 (mGluR5). Primary screening in zebrafish (Danio rerio) revealed a stimulatory effect of two peptides, LCGM-10 and LCGM-15. Target validation studies using calcium ion flux imaging and a luciferase reporter assay confirmed mGluR5 as the target. LCGM-10 showed greater potency than LCGM-15; it was comparable to that of the mGluR5 NAM 2-methyl-6-(phenylethynyl) pyridine (MPEP). Rodent behavioral screening in the open field and elevated plus maze revealed increased locomotor activity in both tests after acute LCGM-10 treatment, supported by further analysis of home cage spontaneous locomotor activity (SLA). The stimulating effect of a single LCGM-10 administration on SLA was evident up to 60 min after administration and was not accompanied by hypokinetic rebound observed for caffeine. According to our results, LCGM-10 has therapeutic potential to treat hypo- and dyskinesias of various etiologies. Further investigation of LCGM-10 effects in the delay discounting model of impulsive choice in rats revealed reduced trait impulsivity after single and chronic administrations, suggesting potential implication for attention deficit hyperactivity disorder, obsessive compulsive disorder, and addictions.
Collapse
|
44
|
Talvio K, Castrén ML. Astrocytes in fragile X syndrome. Front Cell Neurosci 2024; 17:1322541. [PMID: 38259499 PMCID: PMC10800791 DOI: 10.3389/fncel.2023.1322541] [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: 10/16/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Astrocytes have an important role in neuronal maturation and synapse function in the brain. The interplay between astrocytes and neurons is found to be altered in many neurodevelopmental disorders, including fragile X syndrome (FXS) that is the most common inherited cause of intellectual disability and autism spectrum disorder. Transcriptional, functional, and metabolic alterations in Fmr1 knockout mouse astrocytes, human FXS stem cell-derived astrocytes as well as in in vivo models suggest autonomous effects of astrocytes in the neurobiology of FXS. Abnormalities associated with FXS astrocytes include differentiation of central nervous system cell populations, maturation and regulation of synapses, and synaptic glutamate balance. Recently, FXS-specific changes were found more widely in astrocyte functioning, such as regulation of inflammatory pathways and maintenance of lipid homeostasis. Changes of FXS astrocytes impact the brain homeostasis and function both during development and in the adult brain and offer opportunities for novel types of approaches for intervention.
Collapse
Affiliation(s)
| | - Maija L. Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
45
|
Ma M, Yu Q, Delafield DG, Cui Y, Li Z, Li M, Wu W, Shi X, Westmark PR, Gutierrez A, Ma G, Gao A, Xu M, Xu W, Westmark CJ, Li L. On-Tissue Spatial Proteomics Integrating MALDI-MS Imaging with Shotgun Proteomics Reveals Soy Consumption-Induced Protein Changes in a Fragile X Syndrome Mouse Model. ACS Chem Neurosci 2024; 15:119-133. [PMID: 38109073 PMCID: PMC11127747 DOI: 10.1021/acschemneuro.3c00497] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Fragile X syndrome (FXS), the leading cause of inherited intellectual disability and autism, is caused by the transcriptional silencing of the FMR1 gene, which encodes the fragile X messenger ribonucleoprotein (FMRP). FMRP interacts with numerous brain mRNAs that are involved in synaptic plasticity and implicated in autism spectrum disorders. Our published studies indicate that single-source, soy-based diets are associated with increased seizures and autism. Thus, there is an acute need for an unbiased protein marker identification in FXS in response to soy consumption. Herein, we present a spatial proteomics approach integrating mass spectrometry imaging with label-free proteomics in the FXS mouse model to map the spatial distribution and quantify levels of proteins in the hippocampus and hypothalamus brain regions. In total, 1250 unique peptides were spatially resolved, demonstrating the diverse array of peptidomes present in the tissue slices and the broad coverage of the strategy. A group of proteins that are known to be involved in glycolysis, synaptic transmission, and coexpression network analysis suggest a significant association between soy proteins and metabolic and synaptic processes in the Fmr1KO brain. Ultimately, this spatial proteomics work represents a crucial step toward identifying potential candidate protein markers and novel therapeutic targets for FXS.
Collapse
Affiliation(s)
- Min Ma
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Qinying Yu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Daniel G. Delafield
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Yusi Cui
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Zihui Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Miyang Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wenxin Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Xudong Shi
- Division of Otolaryngology, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Alejandra Gutierrez
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Ang Gao
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Meng Xu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
46
|
Gibson JM, Vazquez AH, Yamashiro K, Jakkamsetti V, Ren C, Lei K, Dentel B, Pascual JM, Tsai PT. Cerebellar contribution to autism-relevant behaviors in fragile X syndrome models. Cell Rep 2023; 42:113533. [PMID: 38048226 PMCID: PMC10831814 DOI: 10.1016/j.celrep.2023.113533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 09/01/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
Cerebellar dysfunction has been linked to autism spectrum disorders (ASDs). Although cerebellar pathology has been observed in individuals with fragile X syndrome (FXS) and in mouse models of the disorder, a cerebellar functional contribution to ASD-relevant behaviors in FXS has yet to be fully characterized. In this study, we demonstrate a critical cerebellar role for Fmr1 (fragile X messenger ribonucleoprotein 1) in ASD-relevant behaviors. First, we identify reduced social behaviors, sensory hypersensitivity, and cerebellar dysfunction, with loss of cerebellar Fmr1. We then demonstrate that cerebellar-specific expression of Fmr1 is sufficient to impact social, sensory, cerebellar dysfunction, and cerebro-cortical hyperexcitability phenotypes observed in global Fmr1 mutants. Moreover, we demonstrate that targeting the ASD-implicated cerebellar region Crus1 ameliorates behaviors in both cerebellar-specific and global Fmr1 mutants. Together, these results demonstrate a critical role for the cerebellar contribution to FXS-related behaviors, with implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Jennifer M Gibson
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anthony Hernandez Vazquez
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kunihiko Yamashiro
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vikram Jakkamsetti
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chongyu Ren
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katherine Lei
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Brianne Dentel
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Juan M Pascual
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Peter T Tsai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
47
|
Zou Z, Wei J, Chen Y, Kang Y, Shi H, Yang F, Shi Z, Chen S, Zhou Y, Sepich-Poore C, Zhuang X, Zhou X, Jiang H, Wen Z, Jin P, Luo C, He C. FMRP phosphorylation modulates neuronal translation through YTHDF1. Mol Cell 2023; 83:4304-4317.e8. [PMID: 37949069 PMCID: PMC10872974 DOI: 10.1016/j.molcel.2023.10.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 09/12/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023]
Abstract
RNA-binding proteins (RBPs) control messenger RNA fate in neurons. Here, we report a mechanism that the stimuli-induced neuronal translation is mediated by phosphorylation of a YTHDF1-binding protein FMRP. Mechanistically, YTHDF1 can condense with ribosomal proteins to promote the translation of its mRNA targets. FMRP regulates this process by sequestering YTHDF1 away from the ribosome; upon neuronal stimulation, FMRP becomes phosphorylated and releases YTHDF1 for translation upregulation. We show that a new small molecule inhibitor of YTHDF1 can reverse fragile X syndrome (FXS) developmental defects associated with FMRP deficiency in an organoid model. Our study thus reveals that FMRP and its phosphorylation are important regulators of activity-dependent translation during neuronal development and stimulation and identifies YTHDF1 as a potential therapeutic target for FXS in which developmental defects caused by FMRP depletion could be reversed through YTHDF1 inhibition.
Collapse
Affiliation(s)
- Zhongyu Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Jiangbo Wei
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Yantao Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yunhee Kang
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hailing Shi
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Fan Yang
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Zhuoyue Shi
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Shijie Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Ying Zhou
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Caraline Sepich-Poore
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoxi Zhuang
- Department of Neurobiology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoming Zhou
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hualiang Jiang
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Cheng Luo
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
48
|
D'Antoni S, Schiavi S, Buzzelli V, Giuffrida S, Feo A, Ascone F, Busceti CL, Nicoletti F, Trezza V, Catania MV. Group I and group II metabotropic glutamate receptors are upregulated in the synapses of infant rats prenatally exposed to valproic acid. Psychopharmacology (Berl) 2023; 240:2617-2629. [PMID: 37707611 PMCID: PMC10640443 DOI: 10.1007/s00213-023-06457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
RATIONALE Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction and restricted/stereotyped behavior. Prenatal exposure to valproic acid (VPA) is associated with an increased risk of developing ASD in humans and autistic-like behaviors in rodents. Increasing evidence indicates that dysfunctions of glutamate receptors at synapses are associated with ASD. In the VPA rat model, an involvement of glutamate receptors in autism-like phenotypes has been suggested; however, few studies were carried out on metabotropic glutamate (mGlu) receptors. OBJECTIVES We examined the protein expression levels of group I (mGlu1 and mGlu5) and group II (mGlu2/3) mGlu receptors in rats prenatally exposed to VPA and evaluated the effect of mGlu receptor modulation on an early autism-like phenotype in these animals. METHODS We used western blotting analysis on synaptosomes obtained from forebrain of control and VPA rats at different ages (postnatal day P13, 35, 90) and carried out ultrasonic vocalization (USV) emission test in infant control and VPA rats. RESULTS The expression levels of all these receptors were significantly increased in infant VPA rats. No changes were detected in adolescent and adult rats. An acute treatment with the preferential mGlu2/3 antagonist, LY341495, attenuated the impairment in the USV emission in VPA rats. No effect was observed after a treatment with the mGlu5 selective antagonist, MTEP. CONCLUSIONS Our findings demonstrate that the expression of group I and group II mGlu receptors is upregulated at synapses of infant VPA rats and suggest that mGlu2/3 receptor modulation may have a therapeutic potential in ASD.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Samuele Giuffrida
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Alessandro Feo
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Fabrizio Ascone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | | | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Vincenza Catania
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy.
| |
Collapse
|
49
|
Dziembowska M. How dendritic spines shape is determined by MMP-9 activity in FXS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:171-185. [PMID: 37993177 DOI: 10.1016/bs.irn.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Matrix metalloproteinase-9 (MMP-9) belongs to the family of endopeptidases expressed in neurons and secreted at the synapse in response to neuronal activity. It regulates the pericellular environment by cleaving its protein components. MMP9 is involved in activity-dependent reorganization of spine architecture. In the mouse model of fragile X syndrome (FXS), the most common inherited intellectual disability and the most common single-gene cause of autism, increased synaptic expression of MMP-9 is responsible for the observed dendritic spine abnormalities. In this chapter, I summarize the current data on the molecular regulatory pathways responsible for synaptic MMP-9 expression and discuss the fact that MMP-9 is extracellularly localized, making it a particularly attractive potential target for therapeutic pharmacological intervention in FXS.
Collapse
|
50
|
Li YJ, Zhang K, Sun T, Guo YY, Yang Q, Liu SB, Wu YM, Zhao MG. Improvement of Learning and Memory by Elevating Brain D-Aspartate in a Mouse Model of Fragile X Syndrome. Mol Neurobiol 2023; 60:6410-6423. [PMID: 37453994 PMCID: PMC10533629 DOI: 10.1007/s12035-023-03438-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/10/2023] [Indexed: 07/18/2023]
Abstract
Fragile X syndrome (FXS) is an inherited human mental retardation that arises from expansion of a CGG repeat in the Fmr1 gene, causing loss of the fragile X mental retardation protein (FMRP). It is reported that N-methyl-D-aspartate receptor (NMDAR)-mediated facilitation of long-term potentiation (LTP) and fear memory are impaired in Fmr1 knockout (KO) mice. In this study, biological, pharmacological, and electrophysiological techniques were performed to determine the roles of D-aspartate (D-Asp), a modulator of NMDAR, and its metabolizing enzyme D-aspartate oxidase (DDO) in Fmr1 KO mice. Levels of D-Asp were decreased in the medial prefrontal cortex (mPFC ); however, the levels of its metabolizing enzyme DDO were increased. Electrophysiological recordings indicated that oral drinking of D-Asp recovered LTP induction in mPFC from Fmr1 KO mice. Moreover, chronic oral administration of D-Asp reversed behavioral deficits of cognition and locomotor coordination in Fmr1 KO mice. The therapeutic action of D-Asp was partially through regulating functions of NMDARs and mGluR5/mTOR/4E-BP signaling pathways. In conclusion, supplement of D-Asp may benefit for synaptic plasticity and behaviors in Fmr1 KO mice and offer a potential therapeutic strategy for FXS.
Collapse
Affiliation(s)
- Yu-Jiao Li
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
- Department of Pharmacy, General Hospital of Eastern Theater Command/Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Ting Sun
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Yan-Yan Guo
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Qi Yang
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Ming-Gao Zhao
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China.
| |
Collapse
|