Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Psychiatry ; 29(5): 1392-1405, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38297084

RESUMO

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.


Assuntos
Benzodioxóis , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Neurônios , Proteínas Proto-Oncogênicas c-fyn , Quinases da Família src , Animais , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Camundongos , Quinases da Família src/metabolismo , Benzodioxóis/farmacologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Masculino , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fenótipo , Sinapses/metabolismo , Sinapses/efeitos dos fármacos , Mapas de Interação de Proteínas/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Córtex Cerebral/metabolismo , Córtex Cerebral/efeitos dos fármacos , Quinazolinas
2.
J Biol Chem ; 299(11): 105271, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37741456

RESUMO

The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that acts as a central mediator of translation and plays important roles in cell growth, synaptic plasticity, cancer, and a wide range of developmental disorders. The signaling cascade linking lipid kinases (phosphoinositide 3-kinases), protein kinases (AKT), and translation initiation complexes (EIFs) to mTOR has been extensively modeled, but does not fully describe mTOR system behavior. Here, we use quantitative multiplex coimmunoprecipitation to monitor a protein interaction network (PIN) composed of 300+ binary interactions among mTOR-related proteins. Using a simple model system of serum-deprived or fresh-media-fed mouse 3T3 fibroblasts, we observed extensive PIN remodeling involving 27+ individual protein interactions after 1 h, despite phosphorylation changes observed after only 5 min. Using small molecule inhibitors of phosphoinositide 3-kinase, AKT, mTOR, MEK and ERK, we define subsets of the PIN, termed "modules", that respond differently to each inhibitor. Using primary fibroblasts from individuals with overgrowth disorders caused by pathogenic PIK3CA or MTOR variants, we find that hyperactivation of mTOR pathway components is reflected in a hyperactive PIN. Our data define a "modular" organization of the mTOR PIN in which coordinated groups of interactions respond to the activation or inhibition of distinct nodes, and demonstrate that kinase inhibitors affect the modular network architecture in a complex manner, inconsistent with simple linear models of signal transduction.


Assuntos
Transdução de Sinais , Serina-Treonina Quinases TOR , Animais , Camundongos , Fosforilação , Mapas de Interação de Proteínas , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Células NIH 3T3 , Células Cultivadas , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mutação
3.
J Neurosci ; 41(4): 780-796, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33310753

RESUMO

Huntington disease (HD) is a neurodegenerative disease caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. Therapeutics that lower HTT have shown preclinical promise and are being evaluated in clinical trials. However, clinical assessment of brain HTT lowering presents challenges. We have reported that mutant HTT (mHTT) in the CSF of HD patients correlates with clinical measures, including disease burden as well as motor and cognitive performance. We have also shown that lowering HTT in the brains of HD mice results in correlative reduction of mHTT in the CSF, prompting the use of this measure as an exploratory marker of target engagement in clinical trials. In this study, we investigate the mechanisms of mHTT clearance from the brain in adult mice of both sexes to elucidate the significance of therapy-induced CSF mHTT changes. We demonstrate that, although neurodegeneration increases CSF mHTT concentrations, mHTT is also present in the CSF of mice in the absence of neurodegeneration. Importantly, we show that secretion of mHTT from cells in the CNS followed by glymphatic clearance from the extracellular space contributes to mHTT in the CSF. Furthermore, we observe secretion of wild type HTT from healthy control neurons, suggesting that HTT secretion is a normal process occurring in the absence of pathogenesis. Overall, our data support both passive release and active clearance of mHTT into CSF, suggesting that its treatment-induced changes may represent a combination of target engagement and preservation of neurons.SIGNIFICANCE STATEMENT: Changes in CSF mutant huntingtin (mHTT) are being used as an exploratory endpoint in HTT lowering clinical trials for the treatment of Huntington disease (HD). Recently, it was demonstrated that intrathecal administration of a HTT lowering agent leads to dose-dependent reduction of CSF mHTT in HD patients. However, little is known about how HTT, an intracellular protein, reaches the extracellular space and ultimately the CSF. Our findings that HTT enters CSF by both passive release and active secretion followed by glymphatic clearance may have significant implications for interpretation of treatment-induced changes of CSF mHTT in clinical trials for HD.


Assuntos
Química Encefálica , Proteína Huntingtina/líquido cefalorraquidiano , Doença de Huntington/líquido cefalorraquidiano , Animais , Astrócitos/metabolismo , Biomarcadores/líquido cefalorraquidiano , Feminino , Sistema Glinfático/metabolismo , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Expansão das Repetições de Trinucleotídeos
4.
Mol Psychiatry ; 26(11): 7047-7068, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33888873

RESUMO

Early-onset epileptic encephalopathies are severe disorders often associated with specific genetic mutations. In this context, the CDKL5 deficiency disorder (CDD) is a neurodevelopmental condition characterized by early-onset seizures, intellectual delay, and motor dysfunction. Although crucial for proper brain development, the precise targets of CDKL5 and its relation to patients' symptoms are still unknown. Here, induced pluripotent stem cells derived from individuals deficient in CDKL5 protein were used to generate neural cells. Proteomic and phosphoproteomic approaches revealed disruption of several pathways, including microtubule-based processes and cytoskeleton organization. While CDD-derived neural progenitor cells have proliferation defects, neurons showed morphological alterations and compromised glutamatergic synaptogenesis. Moreover, the electrical activity of CDD cortical neurons revealed hyperexcitability during development, leading to an overly synchronized network. Many parameters of this hyperactive network were rescued by lead compounds selected from a human high-throughput drug screening platform. Our results enlighten cellular, molecular, and neural network mechanisms of genetic epilepsy that could ultimately promote novel therapeutic opportunities for patients.


Assuntos
Síndromes Epilépticas , Animais , Síndromes Epilépticas/genética , Humanos , Camundongos , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases , Proteômica
5.
J Infect Dis ; 222(12): 1965-1973, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32798222

RESUMO

We present a microsphere-based flow cytometry assay that quantifies the ability of plasma to inhibit the binding of spike protein to angiotensin-converting enzyme 2. Plasma from 22 patients who had recovered from mild coronavirus disease 2019 (COVID-19) and expressed anti-spike protein trimer immunoglobulin G inhibited angiotensin-converting enzyme 2-spike protein binding to a greater degree than controls. The degree of inhibition was correlated with anti-spike protein immunoglobulin G levels, neutralizing titers in a pseudotyped lentiviral assay, and the presence of fever during illness. This inhibition assay may be broadly useful to quantify the functional antibody response of patients recovered from COVID-19 or vaccine recipients in a cell-free assay system.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , Testes Sorológicos/métodos , Glicoproteína da Espícula de Coronavírus/imunologia , Adulto , Idoso , Sítios de Ligação , Feminino , Células HEK293 , Humanos , Masculino , Microesferas , Pessoa de Meia-Idade , Plasma/imunologia , Ligação Proteica , SARS-CoV-2/imunologia , Adulto Jovem
6.
J Neurochem ; 146(5): 540-559, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29804286

RESUMO

Cells utilize dynamic, network-level rearrangements in highly interconnected protein interaction networks to transmit and integrate information from distinct signaling inputs. Despite the importance of protein interaction network dynamics, the organizational logic underlying information flow through these networks is not well understood. Previously, we developed the quantitative multiplex co-immunoprecipitation platform, which allows for the simultaneous and quantitative measurement of the amount of co-association between large numbers of proteins in shared complexes. Here, we adapt quantitative multiplex co-immunoprecipitation to define the activity-dependent dynamics of an 18-member protein interaction network in order to better understand the underlying principles governing glutamatergic signal transduction. We first establish that immunoprecipitation detected by flow cytometry can detect activity-dependent changes in two known protein-protein interactions (Homer1-mGluR5 and PSD-95-SynGAP). We next demonstrate that neuronal stimulation elicits a coordinated change in our targeted protein interaction network, characterized by the initial dissociation of Homer1 and SynGAP-containing complexes followed by increased associations among glutamate receptors and PSD-95. Finally, we show that stimulation of distinct glutamate receptor types results in different modular sets of protein interaction network rearrangements, and that cells activate both modules in order to integrate complex inputs. This analysis demonstrates that cells respond to distinct types of glutamatergic input by modulating different combinations of protein co-associations among a targeted network of proteins. Our data support a model of synaptic plasticity in which synaptic stimulation elicits dissociation of pre-existing multiprotein complexes, opening binding slots in scaffold proteins and allowing for the recruitment of additional glutamatergic receptors. Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Assuntos
Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Proteína 4 Homóloga a Disks-Large/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Arcabouço Homer/deficiência , Proteínas de Arcabouço Homer/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/fisiologia , Cloreto de Potássio/farmacologia , Análise de Componente Principal , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
7.
Exp Dermatol ; 25(3): 223-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26660334

RESUMO

Aiming to decipher immunological mechanisms of the autoimmune disorder alopecia areata (AA), we hypothesized that interleukin-6 (IL-6) might be associated with juvenile-onset AA, for which there is currently no experimental model. Upon intramuscular transgenesis to overexpress IL-6 in pregnant female C57BL/6 (B6) mice, we found that the offspring displayed an initial normal and complete juvenile hair growth cycle, but developed alopecia around postnatal day 18. This alopecia was patchy and reversible (non-scarring) and was associated with upregulation of Ulbp1 expression, the only mouse homolog of the human AA-associated ULBP3 gene. Alopecia was also associated with inflammatory infiltration of hair follicles by lymphocytes, including alpha-beta T cells, which contributed to surface hair loss. Despite these apparently shared traits with AA, lesions were dominated by follicular dystrophy that was atypical of human AA disease, sharing some traits consistent with B6 alopecia and dermatitis. Additionally, juvenile-onset alopecia was followed by complete, spontaneous recovery of surface hair, without recurrence of hair loss. Prolonging exposure to IL-6 prolonged the time to recovery, but once recovered, repeating high-dose IL-6 exposure de novo did not re-induce alopecia. These data suggest that although substantial molecular and cellular pathways may be shared, functionally similar alopecia disorders can occur via distinct pathological mechanisms.


Assuntos
Alopecia/genética , Folículo Piloso/fisiopatologia , Interleucina-6/metabolismo , Linfócitos T/citologia , Alopecia/imunologia , Animais , Dermatite/genética , Dermatite/metabolismo , Dermatite/fisiopatologia , Feminino , Inflamação , Interleucina-6/genética , Ligantes , Linfócitos/citologia , Exposição Materna , Camundongos , Camundongos Endogâmicos C57BL , Mães , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Gravidez , Transgenes
8.
Brain Behav Immun ; 48: 139-46, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25816799

RESUMO

Maternal infection during pregnancy increases the risk for neurodevelopmental disorders in offspring. Rodent models have played a critical role in establishing maternal immune activation (MIA) as a causal factor for altered brain and behavioral development in offspring. We recently extended these findings to a species more closely related to humans by demonstrating that rhesus monkeys (Macaca mulatta) prenatally exposed to MIA also develop abnormal behaviors. Here, for the first time, we present initial evidence of underlying brain pathology in this novel nonhuman primate MIA model. Pregnant rhesus monkeys were injected with a modified form of the viral mimic polyI:C (poly ICLC) or saline at the end of the first trimester. Brain tissue was collected from the offspring at 3.5 years and blocks of dorsolateral prefrontal cortex (BA46) were used to analyze neuronal dendritic morphology and spine density using the Golgi-Cox impregnation method. For each case, 10 layer III pyramidal cells were traced in their entirety, including all apical, oblique and basal dendrites, and their spines. We further analyzed somal size and apical dendrite trunk morphology in 30 cells per case over a 30 µm section located 100±10 µm from the soma. Compared to controls, apical dendrites of MIA-treated offspring were smaller in diameter and exhibited a greater number of oblique dendrites. These data provide the first evidence that prenatal exposure to MIA alters dendritic morphology in a nonhuman primate MIA model, which may have profound implications for revealing the underlying neuropathology of neurodevelopmental disorders related to maternal infection.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Neurônios/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Carboximetilcelulose Sódica/análogos & derivados , Carboximetilcelulose Sódica/farmacologia , Forma Celular/efeitos dos fármacos , Feminino , Macaca mulatta , Masculino , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Poli I-C/farmacologia , Polilisina/análogos & derivados , Polilisina/farmacologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia
9.
Exp Dermatol ; 23(4): 272-3, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24588717

RESUMO

Studying signal transduction in skin-resident T cells (sr-T cells) can be limited by the small size of clinical biopsies. Here, we isolated sr-T cells from clinical samples and analysed signalling protein complexes by multiplex immunoprecipitation detected by flow cytometry (mIP-FCM). In samples from two independent donors, antigenic stimulation induced signalling proteins to join shared complexes that were observed in seven pairwise combinations among five proteins. This demonstrates that sr-T cells isolated from small clinical samples provide sufficient material for mIP-FCM-based analysis of signalling-induced protein complexes. We propose that this strategy may be useful for gaining improved mechanistic insight of sr-T cell signal transduction associated with dermatological disease.


Assuntos
Imunoprecipitação/métodos , Complexos Multiproteicos/análise , Pele/química , Linfócitos T/química , Citometria de Fluxo , Humanos , Transdução de Sinais , Pele/citologia , Pele/imunologia
10.
Sci Signal ; 17(826): eadd4671, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38442200

RESUMO

Cells rely on activity-dependent protein-protein interactions to convey biological signals. For chimeric antigen receptor (CAR) T cells containing a 4-1BB costimulatory domain, receptor engagement is thought to stimulate the formation of protein complexes similar to those stimulated by T cell receptor (TCR)-mediated signaling, but the number and type of protein interaction-mediating binding domains differ between CARs and TCRs. Here, we performed coimmunoprecipitation mass spectrometry analysis of a second-generation, CD19-directed 4-1BB:ζ CAR (referred to as bbζCAR) and identified 128 proteins that increased their coassociation after target engagement. We compared activity-induced TCR and CAR signalosomes by quantitative multiplex coimmunoprecipitation and showed that bbζCAR engagement led to the activation of two modules of protein interactions, one similar to TCR signaling that was more weakly engaged by bbζCAR as compared with the TCR and one composed of TRAF signaling complexes that was not engaged by the TCR. Batch-to-batch and interindividual variations in production of the cytokine IL-2 correlated with differences in the magnitude of protein network activation. Future CAR T cell manufacturing protocols could measure, and eventually control, biological variation by monitoring these signalosome activation markers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transdução de Sinais , Antígenos CD19/genética , Membrana Celular , Receptores de Antígenos de Linfócitos T/genética
11.
J Investig Dermatol Symp Proc ; 16(1): S31-3, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24326546

RESUMO

Development of better therapies for the T cell-mediated autoimmune disease alopecia areata (AA) could be expedited by an improved understanding of the immunologic signals underlying its pathogenesis. To approach this, our group is mounting a new technological and analytical platform, multiplex immunoprecipitation detected by flow cytometry (MIF). MIF is designed to allow analysis of collections of protein-protein interactions that participate in T cell signaling webs. Early experiments suggest that MIF can detect the increased protein-protein interaction network activity that occurs under conditions of T cell antigenic stimulation. Future experiments will focus on application of MIF to T cells isolated from AA or control patient samples, to identify critical T cell signaling complexes associated with the disorder.


Assuntos
Alopecia em Áreas/imunologia , Citometria de Fluxo , Imunoprecipitação , Transdução de Sinais , Linfócitos T/metabolismo , Alopecia em Áreas/tratamento farmacológico , Fluorescência , Humanos , Microesferas , Ficoeritrina , Mapas de Interação de Proteínas
12.
bioRxiv ; 2023 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-37577705

RESUMO

The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that acts as a central mediator of translation, and plays important roles in cell growth, synaptic plasticity, cancer, and a wide range of developmental disorders. The signaling cascade linking lipid kinases (PI3Ks), protein kinases (AKT) and translation initiation complexes (EIFs) to mTOR has been extensively modeled, but does not fully describe mTOR system behavior. Here, we use quantitative multiplex co-immunoprecipitation to monitor a protein interaction network (PIN) composed of 300+ binary interactions among mTOR-related proteins. Using a simple model system of serum deprived or fresh-media-fed mouse 3T3 fibroblasts, we observed extensive PIN remodeling involving 27+ individual protein interactions after one hour, despite phosphorylation changes observed after only five minutes. Using small molecule inhibitors of PI3K, AKT, mTOR, MEK and ERK, we define subsets of the PIN, termed 'modules', that respond differently to each inhibitor. Using primary fibroblasts from individuals with overgrowth disorders caused by pathogenic PIK3CA or MTOR variants, we find that hyperactivation of mTOR pathway components is reflected in a hyperactive PIN. Our data define a "modular" organization of the mTOR PIN in which coordinated groups of interactions respond to activation or inhibition of distinct nodes, and demonstrate that kinase inhibitors affect the modular network architecture in a complex manner, inconsistent with simple linear models of signal transduction.

13.
J Neurochem ; 120(4): 611-21, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22122031

RESUMO

Activity-dependent redistribution of ion channels mediates neuronal circuit plasticity and homeostasis, and could provide pro-epileptic or compensatory anti-epileptic responses to a seizure. Thalamocortical neurons transmit sensory information to the cerebral cortex and through reciprocal corticothalamic connections are intensely activated during a seizure. Therefore, we assessed whether a seizure alters ion channel surface expression and consequent neurophysiologic function of thalamocortical neurons. We report a seizure triggers a rapid (<2h) decrease of excitatory postsynaptic current (EPSC)-like current-induced phasic firing associated with increased transient A-type K(+) current. Seizures also rapidly redistributed the A-type K(+) channel subunit Kv4.2 to the neuronal surface implicating a molecular substrate for the increased K(+) current. Glutamate applied in vitro mimicked the effect, suggesting a direct effect of glutamatergic transmission. Importantly, leucine-rich glioma-inactivated-1 (LGI1), a secreted synaptic protein mutated to cause human partial epilepsy, regulated this seizure-induced circuit response. Human epilepsy-associated dominant-negative-truncated mutant LGI1 inhibited the seizure-induced suppression of phasic firing, increase of A-type K(+) current, and recruitment of Kv4.2 surface expression (in vivo and in vitro). The results identify a response of thalamocortical neurons to seizures involving Kv4.2 surface recruitment associated with dampened phasic firing. The results also identify impaired seizure-induced increases of A-type K(+) current as an additional defect produced by the autosomal dominant lateral temporal lobe epilepsy gene mutant that might contribute to the seizure disorder.


Assuntos
Mutação , Inibição Neural/genética , Proteínas/genética , Convulsões/metabolismo , Convulsões/prevenção & controle , Canais de Potássio Shal/antagonistas & inibidores , Canais de Potássio Shal/metabolismo , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Potenciais Pós-Sinápticos Excitadores/genética , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Transgênicos , Condução Nervosa/genética , Neurônios/metabolismo , Neurônios/patologia , Técnicas de Cultura de Órgãos , Transporte Proteico/genética , Proteínas/fisiologia , Convulsões/patologia , Canais de Potássio Shal/genética
14.
Sci Rep ; 12(1): 3207, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217690

RESUMO

Neurons encode information by rapidly modifying synaptic protein complexes, which changes the strength of specific synaptic connections. Homer1 is abundantly expressed at glutamatergic synapses, and is known to alter its binding to metabotropic glutamate receptor 5 (mGlu5) in response to synaptic activity. However, Homer participates in many additional known interactions whose activity-dependence is unclear. Here, we used co-immunoprecipitation and label-free quantitative mass spectrometry to characterize activity-dependent interactions in the cerebral cortex of wildtype and Homer1 knockout mice. We identified a small, high-confidence protein network consisting of mGlu5, Shank2 and 3, and Homer1-3, of which only mGlu5 and Shank3 were significantly reduced following neuronal depolarization. We identified several other proteins that reduced their co-association in an activity-dependent manner, likely mediated by Shank proteins. We conclude that Homer1 dissociates from mGlu5 and Shank3 following depolarization, but our data suggest that direct Homer1 interactions in the cortex may be more limited than expected.


Assuntos
Neurônios , Sinapses , Animais , Córtex Cerebral/metabolismo , Proteínas de Arcabouço Homer/genética , Proteínas de Arcabouço Homer/metabolismo , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo
15.
Cell Rep ; 37(9): 110076, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34852231

RESUMO

A core network of widely expressed proteins within the glutamatergic post-synapse mediates activity-dependent synaptic plasticity throughout the brain, but the specific proteomic composition of synapses differs between brain regions. Here, we address the question, how does proteomic composition affect activity-dependent protein-protein interaction networks (PINs) downstream of synaptic activity? Using quantitative multiplex co-immunoprecipitation, we compare the PIN response of in vivo or ex vivo neurons derived from different brain regions to activation by different agonists or different forms of eyeblink conditioning. We report that PINs discriminate between incoming stimuli using differential kinetics of overlapping and non-overlapping PIN parameters. Further, these "molecular logic rules" differ by brain region. We conclude that although the PIN of the glutamatergic post-synapse is expressed widely throughout the brain, its activity-dependent dynamics show remarkable stimulus-specific and brain-region-specific diversity. This diversity may help explain the challenges in developing molecule-specific drug therapies for neurological disorders.


Assuntos
Piscadela/efeitos dos fármacos , Encéfalo/metabolismo , Metoxi-Hidroxifenilglicol/análogos & derivados , N-Metilaspartato/farmacologia , Mapas de Interação de Proteínas , Proteoma/metabolismo , Sinapses/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Condicionamento Palpebral , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Masculino , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Plasticidade Neuronal , Proteoma/análise , Sinapses/efeitos dos fármacos
16.
bioRxiv ; 2021 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-33851160

RESUMO

Repurposed drugs that block the interaction between the SARS-CoV-2 spike protein and its receptor ACE2 could offer a rapid route to novel COVID-19 treatments or prophylactics. Here, we screened 2701 compounds from a commercial library of drugs approved by international regulatory agencies for their ability to inhibit the binding of recombinant, trimeric SARS-CoV-2 spike protein to recombinant human ACE2. We identified 56 compounds that inhibited binding by <90%, measured the EC 50 of binding inhibition, and computationally modeled the docking of the best inhibitors to both Spike and ACE2. These results highlight an effective screening approach to identify compounds capable of disrupting the Spike-ACE2 interaction as well as identifying several potential inhibitors that could serve as templates for future drug discovery efforts.

17.
Front Pharmacol ; 12: 685308, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34194331

RESUMO

Repurposed drugs that block the interaction between the SARS-CoV-2 spike protein and its receptor ACE2 could offer a rapid route to novel COVID-19 treatments or prophylactics. Here, we screened 2,701 compounds from a commercial library of drugs approved by international regulatory agencies for their ability to inhibit the binding of recombinant, trimeric SARS-CoV-2 spike protein to recombinant human ACE2. We identified 56 compounds that inhibited binding in a concentration-dependent manner, measured the IC50 of binding inhibition, and computationally modeled the docking of the best inhibitors to the Spike-ACE2 binding interface. The best candidates were Thiostrepton, Oxytocin, Nilotinib, and Hydroxycamptothecin with IC50's in the 4-9 µM range. These results highlight an effective screening approach to identify compounds capable of disrupting the Spike-ACE2 interaction, as well as identify several potential inhibitors of the Spike-ACE2 interaction.

18.
Front Mol Neurosci ; 14: 772847, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899181

RESUMO

Patients harboring mutations in the PI3K-AKT-MTOR pathway-encoding genes often develop a spectrum of neurodevelopmental disorders including epilepsy. A significant proportion remains unresponsive to conventional anti-seizure medications. Understanding mutation-specific pathophysiology is thus critical for molecularly targeted therapies. We previously determined that mouse models expressing a patient-related activating mutation in PIK3CA, encoding the p110α catalytic subunit of phosphoinositide-3-kinase (PI3K), are epileptic and acutely treatable by PI3K inhibition, irrespective of dysmorphology. Here we report the physiological mechanisms underlying this dysregulated neuronal excitability. In vivo, we demonstrate epileptiform events in the Pik3ca mutant hippocampus. By ex vivo analyses, we show that Pik3ca-driven hyperactivation of hippocampal pyramidal neurons is mediated by changes in multiple non-synaptic, cell-intrinsic properties. Finally, we report that acute inhibition of PI3K or AKT, but not MTOR activity, suppresses the intrinsic hyperactivity of the mutant neurons. These acute mechanisms are distinct from those causing neuronal hyperactivity in other AKT-MTOR epileptic models and define parameters to facilitate the development of new molecularly rational therapeutic interventions for intractable epilepsy.

19.
Sci Signal ; 14(681)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33947797

RESUMO

Neurons maintain stable levels of excitability using homeostatic synaptic scaling, which adjusts the strength of a neuron's postsynaptic inputs to compensate for extended changes in overall activity. Here, we investigated whether prolonged changes in activity affect network-level protein interactions at the synapse. We assessed a glutamatergic synapse protein interaction network (PIN) composed of 380 binary associations among 21 protein members in mouse neurons. Manipulating the activation of cultured mouse cortical neurons induced widespread bidirectional PIN alterations that reflected rapid rearrangements of glutamate receptor associations involving synaptic scaffold remodeling. Sensory deprivation of the barrel cortex in live mice (by whisker trimming) caused specific PIN rearrangements, including changes in the association between the glutamate receptor mGluR5 and the kinase Fyn. These observations are consistent with emerging models of experience-dependent plasticity involving multiple types of homeostatic responses. However, mice lacking Homer1 or Shank3B did not undergo normal PIN rearrangements, suggesting that the proteins encoded by these autism spectrum disorder-linked genes serve as structural hubs for synaptic homeostasis. Our approach demonstrates how changes in the protein content of synapses during homeostatic plasticity translate into functional PIN alterations that mediate changes in neuron excitability.


Assuntos
Transtorno do Espectro Autista , Plasticidade Neuronal , Animais , Homeostase , Camundongos , Neurônios , Sinapses
20.
Science ; 371(6530)2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33574182

RESUMO

The evolutionarily conserved splicing regulator neuro-oncological ventral antigen 1 (NOVA1) plays a key role in neural development and function. NOVA1 also includes a protein-coding difference between the modern human genome and Neanderthal and Denisovan genomes. To investigate the functional importance of an amino acid change in humans, we reintroduced the archaic allele into human induced pluripotent cells using genome editing and then followed their neural development through cortical organoids. This modification promoted slower development and higher surface complexity in cortical organoids with the archaic version of NOVA1 Moreover, levels of synaptic markers and synaptic protein coassociations correlated with altered electrophysiological properties in organoids expressing the archaic variant. Our results suggest that the human-specific substitution in NOVA1, which is exclusive to modern humans since divergence from Neanderthals, may have had functional consequences for our species' evolution.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Homem de Neandertal/genética , Neurônios/fisiologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Alelos , Processamento Alternativo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Evolução Biológica , Sistemas CRISPR-Cas , Proliferação de Células , Córtex Cerebral/citologia , Regulação da Expressão Gênica no Desenvolvimento , Variação Genética , Genoma , Genoma Humano , Haplótipos , Hominidae/genética , Humanos , Células-Tronco Pluripotentes Induzidas , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Antígeno Neuro-Oncológico Ventral , Organoides , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa