Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 147(1): 29, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38308693

ABSTRACT

The aggregation, mislocalization, and phosphorylation of TDP-43 are pathologic hallmarks of several neurodegenerative diseases and provide a defining criterion for the neuropathologic diagnosis of Limbic-predominant Age-related TDP-43 Encephalopathy (LATE). LATE neuropathologic changes (LATE-NC) are often comorbid with other neurodegenerative pathologies including Alzheimer's disease neuropathologic changes (ADNC). We examined whether TDP-43 regulated cryptic exons accumulate in the hippocampus of neuropathologically confirmed LATE-NC cases. We found that several cryptic RNAs are robustly expressed in LATE-NC cases with or without comorbid ADNC and correlate with pTDP-43 abundance; however, the accumulation of cryptic RNAs is more robust in LATE-NC with comorbid ADNC. Additionally, cryptic RNAs can robustly distinguish LATE-NC from healthy controls and AD cases. These findings expand our current understanding and provide novel potential biomarkers for LATE pathogenesis.


Subject(s)
Alzheimer Disease , Dementia , TDP-43 Proteinopathies , Humans , Brain/pathology , TDP-43 Proteinopathies/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Aging/genetics , Aging/pathology , DNA-Binding Proteins/metabolism , Exons
2.
Sci Adv ; 9(33): eadh0558, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37585521

ABSTRACT

The 1.6-megabase deletion at chromosome 3q29 (3q29Del) is the strongest identified genetic risk factor for schizophrenia, but the effects of this variant on neurodevelopment are not well understood. We interrogated the developing neural transcriptome in two experimental model systems with complementary advantages: isogenic human cortical organoids and isocortex from the 3q29Del mouse model. We profiled transcriptomes from isogenic cortical organoids that were aged for 2 and 12 months, as well as perinatal mouse isocortex, all at single-cell resolution. Systematic pathway analysis implicated dysregulation of mitochondrial function and energy metabolism. These molecular signatures were supported by analysis of oxidative phosphorylation protein complex expression in mouse brain and assays of mitochondrial function in engineered cell lines, which revealed a lack of metabolic flexibility and a contribution of the 3q29 gene PAK2. Together, these data indicate that metabolic disruption is associated with 3q29Del and is conserved across species.


Subject(s)
Intellectual Disability , Neocortex , Schizophrenia , Child , Humans , Animals , Mice , Aged , Schizophrenia/genetics , Chromosome Deletion , Developmental Disabilities/complications , Developmental Disabilities/genetics
3.
bioRxiv ; 2023 May 26.
Article in English | MEDLINE | ID: mdl-36747819

ABSTRACT

Recent advances in the genetics of schizophrenia (SCZ) have identified rare variants that confer high disease risk, including a 1.6 Mb deletion at chromosome 3q29 with a staggeringly large effect size (O.R. > 40). Understanding the impact of the 3q29 deletion (3q29Del) on the developing CNS may therefore lead to insights about the pathobiology of schizophrenia. To gain clues about the molecular and cellular perturbations caused by the 3q29 deletion, we interrogated transcriptomic effects in two experimental model systems with complementary advantages: isogenic human forebrain cortical organoids and isocortex from the 3q29Del mouse model. We first created isogenic lines by engineering the full 3q29Del into an induced pluripotent stem cell line from a neurotypical individual. We profiled transcriptomes from isogenic cortical organoids that were aged for 2 months and 12 months, as well as day p7 perinatal mouse isocortex, all at single cell resolution. Differential expression analysis by genotype in each cell-type cluster revealed that more than half of the differentially expressed genes identified in mouse cortex were also differentially expressed in human cortical organoids, and strong correlations were observed in mouse-human differential gene expression across most major cell-types. We systematically filtered differentially expressed genes to identify changes occurring in both model systems. Pathway analysis on this filtered gene set implicated dysregulation of mitochondrial function and energy metabolism, although the direction of the effect was dependent on developmental timepoint. Transcriptomic changes were validated at the protein level by analysis of oxidative phosphorylation protein complexes in mouse brain tissue. Assays of mitochondrial function in human heterologous cells further confirmed robust mitochondrial dysregulation in 3q29Del cells, and these effects are partially recapitulated by ablation of the 3q29Del gene PAK2 . Taken together these data indicate that metabolic disruption is associated with 3q29Del and is conserved across species. These results converge with data from other rare SCZ-associated variants as well as idiopathic schizophrenia, suggesting that mitochondrial dysfunction may be a significant but overlooked contributing factor to the development of psychotic disorders. This cross-species scRNA-seq analysis of the SCZ-associated 3q29 deletion reveals that this copy number variant may produce early and persistent changes in cellular metabolism that are relevant to human neurodevelopment.

4.
PLoS Pathog ; 18(12): e1010994, 2022 12.
Article in English | MEDLINE | ID: mdl-36508467

ABSTRACT

The emergence of new variants of SARS-CoV-2 necessitates unremitting efforts to discover novel therapeutic monoclonal antibodies (mAbs). Here, we report an extremely potent mAb named P4A2 that can neutralize all the circulating variants of concern (VOCs) with high efficiency, including the highly transmissible Omicron. The crystal structure of the P4A2 Fab:RBD complex revealed that the residues of the RBD that interact with P4A2 are a part of the ACE2-receptor-binding motif and are not mutated in any of the VOCs. The pan coronavirus pseudotyped neutralization assay confirmed that the P4A2 mAb is specific for SARS-CoV-2 and its VOCs. Passive administration of P4A2 to K18-hACE2 transgenic mice conferred protection, both prophylactically and therapeutically, against challenge with VOCs. Overall, our data shows that, the P4A2 mAb has immense therapeutic potential to neutralize the current circulating VOCs. Due to the overlap between the P4A2 epitope and ACE2 binding site on spike-RBD, P4A2 may also be highly effective against a number of future variants.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , COVID-19 , SARS-CoV-2 , Animals , Humans , Mice , Angiotensin-Converting Enzyme 2/chemistry , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , COVID-19/immunology , COVID-19/therapy , Mice, Transgenic , Neutralization Tests , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics
6.
3 Biotech ; 12(9): 202, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35928502

ABSTRACT

We used human semi-synthetic phage antibody gene libraries to select anti-SARS-CoV-2 RBD scFv antibody fragment and subsequent characterization of this novel tetravalent monoclonal antibody targeting conformational epitopes in the receptor binding domain of SARS-CoV-2. Binding studies suggest that II62 tetravalent antibody cross-reacts with RBD protein of SARS-CoV2 and its different variants of concerns. The epitope mapping data reveals that II62 tetravalent antibody targets an epitope that does not directly interferes with RBD: ACE2 interaction. Neutralization studies with live authentic SARS-CoV2 virus suggests that increase in valency of II62 mAb from monovalent to tetravalent doesn't perturbate virus interactions with the ACE2 expressing host cells in cytopathic effect-based (CPE) assay. Supplementary Information: The online version contains supplementary material available at 10.1007/s13205-022-03272-6.

7.
Bio Protoc ; 12(12): e4450, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35864902

ABSTRACT

Phage display is a proven and widely used technology for selecting specific antibodies against desired targets. However, an immense amount of effort is required to identify and screen the desired positive clones from large and diverse combinatorial libraries. On the other hand, the selection of positive binding clones from synthetic and semi-synthetic libraries has an inherent bias toward clones with randomly produced amber stop codons, making it more difficult to identify desirable binding antibodies. To overcome the screening of desired clones with amber codons, we present a step-by-step approach for effective phage library screening to isolate useful antibodies. The procedure calls for creating a simple new vector system for soluble production of phage ELISA positive binding clones with one or more amber stop codons in their single-chain antibody fragment (scFv) gene sequences, which is otherwise difficult in standard screening. Graphical abstract.

8.
Nat Neurosci ; 24(10): 1377-1391, 2021 10.
Article in English | MEDLINE | ID: mdl-34413513

ABSTRACT

Fragile X syndrome (FXS) is caused by the loss of fragile X mental retardation protein (FMRP), an RNA-binding protein that can regulate the translation of specific mRNAs. In this study, we developed an FXS human forebrain organoid model and observed that the loss of FMRP led to dysregulated neurogenesis, neuronal maturation and neuronal excitability. Bulk and single-cell gene expression analyses of FXS forebrain organoids revealed that the loss of FMRP altered gene expression in a cell-type-specific manner. The developmental deficits in FXS forebrain organoids could be rescued by inhibiting the phosphoinositide 3-kinase pathway but not the metabotropic glutamate pathway disrupted in the FXS mouse model. We identified a large number of human-specific mRNAs bound by FMRP. One of these human-specific FMRP targets, CHD2, contributed to the altered gene expression in FXS organoids. Collectively, our study revealed molecular, cellular and electrophysiological abnormalities associated with the loss of FMRP during human brain development.


Subject(s)
Fragile X Syndrome/drug therapy , Fragile X Syndrome/pathology , Neurogenesis/genetics , Prosencephalon/pathology , Adult , Brain/pathology , Cell Differentiation , DNA-Binding Proteins/genetics , Electrophysiological Phenomena , Humans , Male , Models, Neurological , Neurogenesis/drug effects , Neurons/pathology , Phosphatidylinositol 3-Kinases/drug effects , Protein Binding , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/genetics , Receptors, Metabotropic Glutamate/drug effects
9.
Cell Rep ; 35(2): 108991, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852833

ABSTRACT

Transcriptional silencing of the FMR1 gene in fragile X syndrome (FXS) leads to the loss of the RNA-binding protein FMRP. In addition to regulating mRNA translation and protein synthesis, emerging evidence suggests that FMRP acts to coordinate proliferation and differentiation during early neural development. However, whether loss of FMRP-mediated translational control is related to impaired cell fate specification in the developing human brain remains unknown. Here, we use human patient induced pluripotent stem cell (iPSC)-derived neural progenitor cells and organoids to model neurogenesis in FXS. We developed a high-throughput, in vitro assay that allows for the simultaneous quantification of protein synthesis and proliferation within defined neural subpopulations. We demonstrate that abnormal protein synthesis in FXS is coupled to altered cellular decisions to favor proliferative over neurogenic cell fates during early development. Furthermore, pharmacologic inhibition of elevated phosphoinositide 3-kinase (PI3K) signaling corrects both excess protein synthesis and cell proliferation in a subset of patient neural cells.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Biological Assay , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Gene Expression Regulation, Developmental , Humans , Imidazoles/pharmacology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Models, Biological , Morpholines/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neurogenesis/genetics , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Piperazines/pharmacology , Primary Cell Culture , Protein Biosynthesis , Pyrimidinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
10.
Neurobiol Dis ; 145: 105055, 2020 11.
Article in English | MEDLINE | ID: mdl-32829028

ABSTRACT

A GGGGCC hexanucleotide repeat expansion in the first intron of C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Compelling evidence suggests that gain of toxicity from the bidirectionally transcribed repeat expanded RNAs plays a central role in disease pathogenesis. Two potential mechanisms have been proposed including RNA-mediated toxicity and/or the production of toxic dipeptide repeat proteins. In this review, we focus on the role of RNA mediated toxicity in ALS/FTD caused by the C9orf72 mutation and discuss arguments for and against this mechanism. In addition, we summarize how G4C2 repeat RNAs can elicit toxicity and potential therapeutic strategies to mitigate RNA-mediated toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , Frontotemporal Dementia/pathology , RNA/toxicity , Amyotrophic Lateral Sclerosis/genetics , Animals , DNA Repeat Expansion , Humans
11.
Mol Autism ; 11(1): 52, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32560741

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS), a neurodevelopmental disorder, is a leading monogenetic cause of intellectual disability and autism spectrum disorder. Notwithstanding the extensive studies using rodent and other pre-clinical models of FXS, which have provided detailed mechanistic insights into the pathophysiology of this disorder, it is only relatively recently that human stem cell-derived neurons have been employed as a model system to further our understanding of the pathophysiological events that may underlie FXS. Our study assesses the physiological properties of human pluripotent stem cell-derived cortical neurons lacking fragile X mental retardation protein (FMRP). METHODS: Electrophysiological whole-cell voltage- and current-clamp recordings were performed on two control and three FXS patient lines of human cortical neurons derived from induced pluripotent stem cells. In addition, we also describe the properties of an isogenic pair of lines in one of which FMR1 gene expression has been silenced. RESULTS: Neurons lacking FMRP displayed bursts of spontaneous action potential firing that were more frequent but shorter in duration compared to those recorded from neurons expressing FMRP. Inhibition of large conductance Ca2+-activated K+ currents and the persistent Na+ current in control neurons phenocopies action potential bursting observed in neurons lacking FMRP, while in neurons lacking FMRP pharmacological potentiation of voltage-dependent Na+ channels phenocopies action potential bursting observed in control neurons. Notwithstanding the changes in spontaneous action potential firing, we did not observe any differences in the intrinsic properties of neurons in any of the lines examined. Moreover, we did not detect any differences in the properties of miniature excitatory postsynaptic currents in any of the lines. CONCLUSIONS: Pharmacological manipulations can alter the action potential burst profiles in both control and FMRP-null human cortical neurons, making them appear like their genetic counterpart. Our studies indicate that FMRP targets that have been found in rodent models of FXS are also potential targets in a human-based model system, and we suggest potential mechanisms by which activity is altered.


Subject(s)
Action Potentials/physiology , Cerebral Cortex/pathology , Fragile X Mental Retardation Protein/metabolism , Induced Pluripotent Stem Cells/pathology , Neurons/pathology , Action Potentials/drug effects , Adolescent , Animals , Cell Differentiation/drug effects , Child, Preschool , Humans , Indoles/pharmacology , Induced Pluripotent Stem Cells/drug effects , Male , Mice , Neurons/drug effects , Riluzole/pharmacology , Sodium Channels/metabolism , Veratridine/pharmacology , Young Adult
12.
Elife ; 92020 06 08.
Article in English | MEDLINE | ID: mdl-32510328

ABSTRACT

The sorting of RNA molecules to subcellular locations facilitates the activity of spatially restricted processes. We have analyzed subcellular transcriptomes of FMRP-null mouse neuronal cells to identify transcripts that depend on FMRP for efficient transport to neurites. We found that these transcripts contain an enrichment of G-quadruplex sequences in their 3' UTRs, suggesting that FMRP recognizes them to promote RNA localization. We observed similar results in neurons derived from Fragile X Syndrome patients. We identified the RGG domain of FMRP as important for binding G-quadruplexes and the transport of G-quadruplex-containing transcripts. Finally, we found that the translation and localization targets of FMRP were distinct and that an FMRP mutant that is unable to bind ribosomes still promoted localization of G-quadruplex-containing messages. This suggests that these two regulatory modes of FMRP may be functionally separated. These results provide a framework for the elucidation of similar mechanisms governed by other RNA-binding proteins.


Subject(s)
Fragile X Mental Retardation Protein , Neurites/metabolism , Neurons/metabolism , RNA Transport/genetics , RNA, Messenger , Animals , Cells, Cultured , Fragile X Mental Retardation Protein/chemistry , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome , G-Quadruplexes , Gene Knockout Techniques , Mice , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Neuron ; 107(2): 292-305.e6, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32375063

ABSTRACT

GGGGCC hexanucleotide repeat expansions (HREs) in C9orf72 cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and lead to the production of aggregating dipeptide repeat proteins (DPRs) via repeat associated non-AUG (RAN) translation. Here, we show the similar intronic GGCCTG HREs that causes spinocerebellar ataxia type 36 (SCA36) is also translated into DPRs, including poly(GP) and poly(PR). We demonstrate that poly(GP) is more abundant in SCA36 compared to c9ALS/FTD patient tissue due to canonical AUG-mediated translation from intron-retained GGCCTG repeat RNAs. However, the frequency of the antisense RAN translation product poly(PR) is comparable between c9ALS/FTD and SCA36 patient samples. Interestingly, in SCA36 patient tissue, poly(GP) exists as a soluble species, and no TDP-43 pathology is present. We show that aggregate-prone chimeric DPR (cDPR) species underlie the divergent DPR pathology between c9ALS/FTD and SCA36. These findings reveal key differences in translation, solubility, and protein aggregation of DPRs between c9ALS/FTD and SCA36.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Dipeptides/genetics , Frontotemporal Dementia/genetics , Mutant Chimeric Proteins/genetics , Spinocerebellar Ataxias/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Antisense Elements (Genetics)/genetics , DNA Repeat Expansion , Female , Humans , Introns/genetics , Mice , Mice, Inbred C57BL , Pregnancy , Repetitive Sequences, Nucleic Acid
14.
Brain Res ; 1693(Pt A): 24-36, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29653083

ABSTRACT

Research in the past decades has unfolded the multifaceted role of Fragile X mental retardation protein (FMRP) and how its absence contributes to the pathophysiology of Fragile X syndrome (FXS). Excess signaling through group 1 metabotropic glutamate receptors is commonly observed in mouse models of FXS, which in part is attributed to dysregulated translation and downstream signaling. Considering the wide spectrum of cellular and physiologic functions that loss of FMRP can affect in general, it may be advantageous to pursue disease mechanism based treatments that directly target translational components or signaling factors that regulate protein synthesis. Various FMRP targets upstream and downstream of the translational machinery are therefore being investigated to further our understanding of the molecular mechanism of RNA and protein synthesis dysregulation in FXS as well as test their potential role as therapeutic interventions to alleviate FXS associated symptoms. In this review, we will broadly discuss recent advancements made towards understanding the role of FMRP in translation regulation, new pre-clinical animal models with FMRP targets located at different levels of the translational and signal transduction pathways for therapeutic intervention as well as future use of stem cells to model FXS associated phenotypes.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/genetics , Animals , Dendrites/metabolism , Disease Models, Animal , Fragile X Syndrome/physiopathology , Gene Expression Regulation , Humans , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA, Messenger/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction
15.
Discoveries (Craiova) ; 6(4): e83, 2018 Dec 31.
Article in English | MEDLINE | ID: mdl-32309603

ABSTRACT

Background and Objectives: Gastric adenocarcinoma is one of the most common malignant tumors and a major cause of cancer death worldwide, especially in developing countries. Her2/neu gene amplification and protein overexpression in breast cancer is a golden criterion for the targeted therapy with trastuzumab. However, the role of Her2 as a prognostic factor in gastric cancer is still controversial. The purpose of this study was to evaluate the frequency of Her2 oncogene overexpression and concordance between the results for Her2 protein expression and gene amplification. Materials and Methods: A total of 65 retroprospective cases with gastric adenocarcinoma, including biopsy and resected specimens obtained between July 2015 to December 2017, were analyzed. Her2/neu expression was determined by Immuno-histochemistry (IHC). Equivocal and some selected cases were submitted for FISH to detect Her2/neu gene amplification. Results: In the present study, out of 65 patients of gastric adenocarcinoma, there were 50 males and 15 females, with mean age of 54.52 years. The majority of tumors were located within the antropyloric region. We found 27 (41.4%) positivity, scored as IHC 3+ and IHC 2+, and 38 (58.3%) negativity, scored as IHC 1+ and IHC 0. We also evidentiated a significant difference between Her2/neu expression with age (p=0.010) and depth of invasion (p=0.020).Her2/neu gene was amplified only in 13 cases, 4 cases were of Her2/neu (3+) positive, 11 cases (39.3%) Her2/neu (2+) with IHC staining. The concordance rate between the results of IHC and FISH in all 18 cases was 83.3%. Conclusion: IHC detection can be carried out to guide the treatment when FISH detection cannot be performed. Overexpression of Her 2/neu in gastric adenocarcinoma could potentially be used in selecting the patients who can get benefit from the anti-Her2/neu targeted therapy.

16.
J Neurosci ; 35(19): 7643-53, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972187

ABSTRACT

Dysbindin is a schizophrenia susceptibility factor and subunit of the biogenesis of lysosome-related organelles complex 1 (BLOC-1) required for lysosome-related organelle biogenesis, and in neurons, synaptic vesicle assembly, neurotransmission, and plasticity. Protein networks, or interactomes, downstream of dysbindin/BLOC-1 remain partially explored despite their potential to illuminate neurodevelopmental disorder mechanisms. Here, we conducted a proteome-wide search for polypeptides whose cellular content is sensitive to dysbindin/BLOC-1 loss of function. We identified components of the vesicle fusion machinery as factors downregulated in dysbindin/BLOC-1 deficiency in neuroectodermal cells and iPSC-derived human neurons, among them the N-ethylmaleimide-sensitive factor (NSF). Human dysbindin/BLOC-1 coprecipitates with NSF and vice versa, and both proteins colocalized in a Drosophila model synapse. To test the hypothesis that NSF and dysbindin/BLOC-1 participate in a pathway-regulating synaptic function, we examined the role for NSF in dysbindin/BLOC-1-dependent synaptic homeostatic plasticity in Drosophila. As previously described, we found that mutations in dysbindin precluded homeostatic synaptic plasticity elicited by acute blockage of postsynaptic receptors. This dysbindin mutant phenotype is fully rescued by presynaptic expression of either dysbindin or Drosophila NSF. However, neither reduction of NSF alone or in combination with dysbindin haploinsufficiency impaired homeostatic synaptic plasticity. Our results demonstrate that dysbindin/BLOC-1 expression defects result in altered cellular content of proteins of the vesicle fusion apparatus and therefore influence synaptic plasticity.


Subject(s)
Drosophila Proteins/metabolism , Dystrophin-Associated Proteins/metabolism , Gene Expression Regulation/physiology , N-Ethylmaleimide-Sensitive Proteins/metabolism , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Drosophila , Drosophila Proteins/genetics , Dysbindin , Dystrophin-Associated Proteins/genetics , Gene Expression Regulation/genetics , Humans , Melanoma/pathology , N-Ethylmaleimide-Sensitive Proteins/genetics , Nerve Net/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , SNARE Proteins/metabolism , Synapses/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
17.
Cell Rep ; 11(5): 681-8, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921527

ABSTRACT

Distinct isoforms of the PI3K catalytic subunit have specialized functions in the brain, but their role in cognition is unknown. Here, we show that the catalytic subunit p110ß plays an important role in prefrontal cortex (PFC)-dependent cognitive defects in mouse models of Fragile X syndrome (FXS), an inherited intellectual disability. FXS is caused by loss of function of the fragile X mental retardation protein (FMRP), which binds and translationally represses mRNAs. PFC-selective knockdown of p110ß, an FMRP target that is translationally upregulated in FXS, reverses deficits in higher cognition in Fmr1 knockout mice. Genetic full-body reduction of p110ß in Fmr1 knockout mice normalizes excessive PI3K activity, restores stimulus-induced protein synthesis, and corrects increased dendritic spine density and behavior. Notably, adult-onset PFC-selective Fmr1 knockdown mice show impaired cognition, which is rescued by simultaneous p110ß knockdown. Our results suggest that FMRP-mediated control of p110ß is crucial for neuronal protein synthesis and cognition.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Cognition/physiology , Fragile X Syndrome/pathology , Animals , Behavior, Animal , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Dendritic Spines/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Male , Mice , Mice, Knockout , Prefrontal Cortex/metabolism , Protein Biosynthesis , Protein Structure, Tertiary , Receptor, Metabotropic Glutamate 5/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...