Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; : e00376, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38876822

ABSTRACT

The neurodevelopmental disorder Pitt Hopkins syndrome (PTHS) causes clinical symptoms similar to Rett syndrome (RTT) patients. However, RTT is caused by MECP2 mutations whereas mutations in the TCF4 gene lead to PTHS. The mechanistic commonalities underling these two disorders are unknown, but their shared symptomology suggest that convergent pathway-level disruption likely exists. We reprogrammed patient skin derived fibroblasts into induced neuronal progenitor cells. Interestingly, we discovered that MeCP2 levels were decreased in PTHS patient iNPCs relative to healthy controls and that both iNPCs and iAstrocytes displayed defects in function and differentiation in a mutation-specific manner. When Tcf4+/- mice were genetically crossed with mice overexpressing MeCP2, molecular and phenotypic defects were significantly ameliorated, underlining and important role of MeCP2 in PTHS pathology. Importantly, post-natal intracerebroventricular gene replacement therapy with adeno-associated viral vector serotype 9 (AAV9)-expressing MeCP2 (AAV9.P546.MeCP2) significantly improved iNPC and iAstrocyte function and effectively ameliorated histological and behavioral defects in Tcf4+/- mice. Combined, our data suggest a previously unknown role of MeCP2 in PTHS pathology and common pathways that might be affected in multiple neurodevelopmental disorders. Our work highlights potential novel therapeutic targets for PTHS, including upregulation of MeCP2 expression or its downstream targets or, potentially, MeCP2-based gene therapy.

2.
Ann Clin Transl Neurol ; 10(10): 1790-1801, 2023 10.
Article in English | MEDLINE | ID: mdl-37545094

ABSTRACT

OBJECTIVE: Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder caused by autosomal-dominant pathogenic variants in either the TSC1 or TSC2 gene, and it is characterized by hamartomas in multiple organs, such as skin, kidney, lung, and brain. These changes can result in epilepsy, learning disabilities, and behavioral complications, among others. The mechanistic link between TSC and the mechanistic target of the rapamycin (mTOR) pathway is well established, thus mTOR inhibitors can potentially be used to treat the clinical manifestations of the disorder, including epilepsy. METHODS: In this study, we tested the efficacy of a novel mTOR catalytic inhibitor (here named Tool Compound 1 or TC1) previously reported to be more brain-penetrant compared with other mTOR inhibitors. Using a well-characterized hypomorphic Tsc2 mouse model, which displays a translationally relevant seizure phenotype, we tested the efficacy of TC1. RESULTS: Our results show that chronic treatment with this novel mTOR catalytic inhibitor (TC1), which affects both the mTORC1 and mTORC2 signaling complexes, reduces seizure burden, and extends the survival of Tsc2 hypomorphic mice, restoring species typical weight gain over development. INTERPRETATION: Novel mTOR catalytic inhibitor TC1 exhibits a promising therapeutic option in the treatment of TSC.


Subject(s)
Epilepsy , Tuberous Sclerosis , Mice , Animals , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tumor Suppressor Proteins/genetics , MTOR Inhibitors , TOR Serine-Threonine Kinases/genetics , Disease Models, Animal , Epilepsy/genetics , Seizures/drug therapy
3.
Ann Clin Transl Neurol ; 10(9): 1695-1699, 2023 09.
Article in English | MEDLINE | ID: mdl-37452008

ABSTRACT

Dravet syndrome (DS) is a monogenic, often refractory, epilepsy resultant from SCN1A haploinsufficiency in humans. A novel therapeutic target in DS that can be engaged in isolation or as adjunctive therapy is highly desirable. Here, we demonstrate reduced expression of the rodent glutamate transporter type 1 (GLT-1) in a DS mouse model, and in wild type mouse strains where Scn1a haploinsufficiency is most likely to cause epilepsy, indicating that GLT-1 depression may play a role in DS seizures. As GLT-1 can be upregulated by common and safe FDA-approved medications, this strategy may be an attractive, viable, and novel avenue for DS treatment.


Subject(s)
Epilepsies, Myoclonic , Epilepsy , Excitatory Amino Acid Transporter 2 , Animals , Humans , Mice , Amino Acid Transport System X-AG , Epilepsies, Myoclonic/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism
4.
Neurotherapeutics ; 19(4): 1340-1352, 2022 07.
Article in English | MEDLINE | ID: mdl-35670902

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder that is characterized by developmental regression, loss of communicative ability, stereotyped hand wringing, cognitive impairment, and central apneas, among many other symptoms. RTT is caused by loss-of-function mutations in a methyl-reader known as methyl-CpG-binding protein 2 (MeCP2), a protein that links epigenetic changes on DNA to larger chromatin structure. Historically, target identification for RTT has relied heavily on Mecp2 knockout mice; however, we recently adopted the alternative approach of performing transcriptional profiling in autopsy samples from RTT patients. Through this mechanism, we identified muscarinic acetylcholine receptors (mAChRs) as potential therapeutic targets. Here, we characterized a cohort of 40 temporal cortex samples from individuals with RTT and quantified significantly decreased levels of the M1, M2, M3, and M5 mAChRs subtypes relative to neurotypical controls. Of these four subtypes, M1 expression demonstrated a linear relationship with MeCP2 expression, such that M1 levels were only diminished in contexts where MeCP2 was also significantly decreased. Further, we show that M1 potentiation with the positive allosteric modulator (PAM) VU0453595 (VU595) rescued social preference, spatial memory, and associative memory deficits, as well as decreased apneas in Mecp2+/- mice. VU595's efficacy on apneas in Mecp2+/- mice was mediated by the facilitation of the transition from inspiration to expiration. Molecular analysis correlated rescue with normalized global gene expression patterns in the brainstem and hippocampus, as well as increased Gsk3ß inhibition and NMDA receptor trafficking. Together, these data suggest that M1 PAMs could represent a new class of RTT therapeutics.


Subject(s)
Rett Syndrome , Mice , Animals , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Rett Syndrome/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Apnea , Receptors, N-Methyl-D-Aspartate , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Knockout , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Chromatin
5.
Neuropharmacology ; 209: 109022, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35248529

ABSTRACT

Rett syndrome (RTT) and MECP2 Duplication syndrome (MDS) have opposing molecular origins in relation to expression and function of the transcriptional regulator Methyl-CpG-binding protein 2 (MeCP2). Several clinical and preclinical phenotypes, however, are shared between these disorders. Modulation of MeCP2 levels has recently emerged as a potential treatment option for both of these diseases. However, toxicity concerns remain with these approaches. Here, we focus on pharmacologically modulating the group II metabotropic glutamate receptors (mGlu), mGlu2 and mGlu3, which are two downstream targets of MeCP2 that are bidirectionally affected in expression in RTT patients and mice (Mecp2Null/+) versus an MDS mouse model (MECP2Tg1/o). Mecp2Null/+ and MECP2Tg1/o animals also exhibit contrasting phenotypes in trace fear acquisition, a form of temporal associative learning and memory, with trace fear deficiency observed in Mecp2Null/+ mice and abnormally enhanced trace fear acquisition in MECP2Tg1/o animals. In Mecp2Null/+ mice, treatment with the mGlu2/3 agonist LY379268 reverses the deficit in trace fear acquisition, and mGlu2/3 antagonism with LY341495 normalizes the abnormal trace fear learning and memory phenotype in MECP2Tg1/o mice. Altogether, these data highlight the role of group II mGlu receptors in RTT and MDS and demonstrate that both mGlu2 and mGlu3 may be potential therapeutic targets for these disorders.


Subject(s)
Mental Retardation, X-Linked , Receptors, Metabotropic Glutamate , Rett Syndrome , Animals , Disease Models, Animal , Humans , Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/metabolism , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Rett Syndrome/metabolism
6.
Genes Brain Behav ; 21(1): e12752, 2022 01.
Article in English | MEDLINE | ID: mdl-34002468

ABSTRACT

De novo loss-of-function mutations in methyl-CpG-binding protein 2 (MeCP2) lead to the neurodevelopmental disorder Rett syndrome (RTT). Despite promising results from strategies aimed at increasing MeCP2 levels, additional studies exploring how hypomorphic MeCP2 mutations impact the therapeutic window are needed. Here, we investigated the consequences of genetically introducing a wild-type MECP2 transgene in the Mecp2 R133C mouse model of RTT. The MECP2 transgene reversed the majority of RTT-like phenotypes exhibited by male and female Mecp2 R133C mice. However, three core symptom domains were adversely affected in female Mecp2R133C/+ animals; these phenotypes resemble those observed in disease contexts of excess MeCP2. Parallel control experiments in Mecp2Null/+ mice linked these adverse effects to the hypomorphic R133C mutation. Collectively, these data provide evidence regarding the safety and efficacy of genetically overexpressing functional MeCP2 in Mecp2 R133C mice and suggest that personalized approaches may warrant consideration for the clinical assessment of MeCP2-targeted therapies.


Subject(s)
Genetic Therapy/methods , Methyl-CpG-Binding Protein 2/genetics , Phenotype , Rett Syndrome/therapy , Animals , Female , Male , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mutation , Rett Syndrome/genetics
7.
Neuropsychopharmacology ; 46(12): 2148-2157, 2021 11.
Article in English | MEDLINE | ID: mdl-34035469

ABSTRACT

Clinical and translational studies suggest that prefrontal cortex (PFC) dysregulation is a hallmark feature of several affective disorders. Thus, investigating the mechanisms involved in the regulation of PFC function and synaptic plasticity could aid in developing new medications. In recent years, the mGlu2 and mGlu3 subtypes of metabotropic glutamate (mGlu) receptors have emerged as exciting potential targets for the treatment of affective disorders, as mGlu2/3 antagonists exert antidepressant-like effects across many rodent models. Several recent studies suggest that presynaptic mGlu2 receptors may contribute to these effects by regulating excitatory transmission at synapses from the thalamus to the PFC. Interestingly, we found that mGlu3 receptors also inhibit excitatory drive to the PFC but act by inducing long-term depression (LTD) at amygdala-PFC synapses. It remains unclear, however, whether blockade of presynaptic, postsynaptic, or glial mGlu3 receptors contribute to long-term effects on PFC circuit function and antidepressant-like effects of mGlu2/3 antagonists. To address these outstanding questions, we leveraged transgenic Grm3fl/fl mice and viral-mediated gene transfer to genetically ablate mGlu3 receptors from pyramidal cells in the frontal cortex of adult mice of all sexes. Consistent with a role for mGlu3 in PFC pyramidal cells, mGlu3-dependent amygdala-cortical LTD was eliminated following mGlu3 receptor knockdown. Furthermore, knockdown mice displayed a modest, task-specific anxiolytic phenotype and decreased passive coping behaviors. These studies reveal that postsynaptic mGlu3 receptors are critical for mGlu3-dependent LTD and provide convergent genetic evidence suggesting that modulating cortical mGlu3 receptors may provide a promising new approach for the treatment of mood disorders.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate/genetics , Animals , Mice , Neuronal Plasticity , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism
8.
ACS Chem Neurosci ; 9(9): 2210-2217, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29227625

ABSTRACT

Rett syndrome and MECP2 Duplication syndrome are neurodevelopmental disorders attributed to loss-of-function mutations in, or duplication of, the gene encoding methyl-CpG-binding protein 2 (MeCP2), respectively. We recently reported decreased expression and function of the metabotropic glutamate receptor 7 (mGlu7) in a mouse model of Rett syndrome. Positive allosteric modulation of mGlu7 activity was sufficient to improve several disease phenotypes including cognition. Here, we tested the hypothesis that mGlu7 expression would be reciprocally regulated in a mouse model of MECP2 Duplication syndrome, such that negative modulation of mGlu7 activity would exert therapeutic benefit. To the contrary, we report that mGlu7 is not functionally increased in mice overexpressing MeCP2 and that neither genetic nor pharmacological reduction of mGlu7 activity impacts phenotypes that are antiparallel to those observed in Rett syndrome model mice. These data expand our understanding of how mGlu7 expression and function is affected by changes in MeCP2 dosage and have important implications for the therapeutic development of mGlu7 modulators.


Subject(s)
Anxiety/metabolism , Fear/physiology , Learning/physiology , Mental Retardation, X-Linked/genetics , Receptors, Metabotropic Glutamate/genetics , Allosteric Regulation , Animals , Anxiety/physiopathology , Anxiety/psychology , Benzoxazoles/pharmacology , Disease Models, Animal , Excitatory Amino Acid Agonists/pharmacology , Fear/drug effects , Fear/psychology , Learning/drug effects , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Mice , Phenotype , Phosphinic Acids/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism
9.
J Nanosci Nanotechnol ; 16(7): 7720-7730, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27547159

ABSTRACT

Noroviruses (NoV) are the leading cause of nonbacterial gastroenteritis in humans, and replicate extensively in the human gastrointestinal (GI) tract. Silica (also known as silicon dioxide, SiO2) nanoparticles (NPs) used in processed foods, dairy products, and beverages also accumulate in the GI tract. We investigated the effect of silica NPs on NoV replication and host cell response during virus infection, using murine norovirus (MNV-1) infection of RAW 264.7 murine macrophages. Pretreatment with 10 µg/ml silica significantly reduced the viability of macrophages, but no cumulative effects on viability of macrophages were observed with MNV-1 infection. No difference was observed between exposure to control or silica NPs on either the quantity of viral genome copies or the production of infectious virus in macrophages infected with MNV-1. Silica NPs reduced the ability of macrophages to upregulate genes encoding bone morphogenic proteins (BMPs), chemokine ligands and cytokines for which expression levels were otherwise found to be upregulated in response to MNV-1 infection. Furthermore, silica NPs reduced the levels of proinflammatory cytokines secreted by macrophages in response to MNV infection. Finally, silica NPs with MNV-1 infection produced a genotoxic insult to macrophages. Strikingly, this genotoxic insult was also found to occur as a synergistic effect of silica NPs and feline calicivirus infection in feline kidney epithelial cells. Taken together, our study suggests important safety considerations related to reducing exposure to silica NPs affecting the GI tract in individuals infected with NoVs and possibly other foodborne viruses.

10.
Placenta ; 44: 69-79, 2016 08.
Article in English | MEDLINE | ID: mdl-27452440

ABSTRACT

INTRODUCTION: Infection accounts for over 40% of preterm premature rupture of the fetal membranes (PPROM), a major cause of preterm birth. Toll-like receptors (TLR) play key roles in pathogen surveillance but their expression and function in amnion mesenchymal cells (AMC) is unclear. The aims of this study were to determine the expression of all TLR isoforms and the effect of macrophage-activating lipoprotein-2 (MALP-2), derived from a common pathogen involved in PPROM, on human AMC. METHODS: AMC were isolated from normal, term amnion from repeat cesarean section. Semi-quantitative RT-PCR, immunocytochemistry, immunohistochemistry and western blotting were used to detect TLR isoform expression. Immunocytochemistry of NF-κB p65, pro-inflammatory cytokine secretion (ELISA), MTT assay, LDH assay, immunoblotting of cytosolic cytochrome c and cleaved caspase-3, and expression of 84 microRNAs by Qiagen miRNA PCR array were used to determine the functional effect of MALP-2 on AMC. RESULTS: TLR1-10 was detected in AMC, and protein expression of TLR2, 4, and 6 were confirmed. MALP-2 induced nuclear translocation of p65, reaching significance after 45 min (ANOVA, P < 0.05). MALP-2 did not cause apoptosis but did lead to significant secretion of IL-4, IL-6, and IL-8 (P < 0.05, 0.01, 0.001, respectively) and significant changes in miRNA-320a and miRNA-18a (P < 0.05). DISCUSSION: These results suggest that AMC elicit a pro-inflammatory response following stimulation with the known TLR2/6 ligand MALP-2. This data supports the idea that AMC express the innate immune system receptors that could help with immune surveillance during infection and contribute to inflammatory responses that lead to PPROM.


Subject(s)
Amnion/metabolism , Cytokines/metabolism , Inflammation/metabolism , Lipopeptides/pharmacology , Toll-Like Receptors/metabolism , Amnion/cytology , Amnion/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Female , Humans , Pregnancy , Signal Transduction/drug effects , Toll-Like Receptors/genetics , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...