Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 22(5): e3002596, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38718086

RESUMO

Autism spectrum disorders (ASD) frequently accompany macrocephaly, which often involves hydrocephalic enlargement of brain ventricles. Katnal2 is a microtubule-regulatory protein strongly linked to ASD, but it remains unclear whether Katnal2 knockout (KO) in mice leads to microtubule- and ASD-related molecular, synaptic, brain, and behavioral phenotypes. We found that Katnal2-KO mice display ASD-like social communication deficits and age-dependent progressive ventricular enlargements. The latter involves increased length and beating frequency of motile cilia on ependymal cells lining ventricles. Katnal2-KO hippocampal neurons surrounded by enlarged lateral ventricles show progressive synaptic deficits that correlate with ASD-like transcriptomic changes involving synaptic gene down-regulation. Importantly, early postnatal Katnal2 re-expression prevents ciliary, ventricular, and behavioral phenotypes in Katnal2-KO adults, suggesting a causal relationship and a potential treatment. Therefore, Katnal2 negatively regulates ependymal ciliary function and its deletion in mice leads to ependymal ciliary hyperfunction and hydrocephalus accompanying ASD-related behavioral, synaptic, and transcriptomic changes.


Assuntos
Transtorno do Espectro Autista , Cílios , Epêndima , Camundongos Knockout , Fenótipo , Animais , Masculino , Camundongos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Comportamento Animal , Cílios/metabolismo , Modelos Animais de Doenças , Epêndima/metabolismo , Hipocampo/metabolismo , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Hidrocefalia/fisiopatologia , Katanina/metabolismo , Katanina/genética , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Sinapses/metabolismo , Transcriptoma/genética
2.
Mol Cell ; 72(3): 426-443.e12, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30401430

RESUMO

Protein phase separation by low-complexity, intrinsically disordered domains generates membraneless organelles and links to neurodegeneration. Cellular prion protein (PrPC) contains such domains, causes spongiform degeneration, and is a receptor for Alzheimer's amyloid-ß oligomers (Aßo). Here, we show that PrPC separates as a liquid phase, in which α-helical Thr become unfolded. At the cell surface, PrPC Lys residues interact with Aßo to create a hydrogel containing immobile Aßo and relatively mobile PrPC. The Aßo/PrP hydrogel has a well-defined stoichiometry and dissociates with excess Aßo. NMR studies of hydrogel PrPC reveal a distinct α-helical conformation for natively unfolded amino-terminal Gly and Ala residues. Aßo/PrP hydrogel traps signal-transducing mGluR5 on the plasma membrane. Recombinant PrPC extracts endogenous Aßo from human Alzheimer's soluble brain lysates into hydrogel, and a PrPC antagonist releases Aßo from endogenous brain hydrogel. Thus, coupled phase and conformational transitions of PrPC are driven by Aß species from Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Proteínas PrPC/química , Proteínas PrPC/fisiologia , Doença de Alzheimer/metabolismo , Animais , Encéfalo , Células COS , Linhagem Celular , Membrana Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Hidrogéis , Imageamento por Ressonância Magnética/métodos , Conformação Molecular , Neurônios , Príons/química , Príons/fisiologia , Ligação Proteica , Receptor de Glutamato Metabotrópico 5 , Transdução de Sinais
3.
J Am Chem Soc ; 146(20): 13846-13853, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38652033

RESUMO

Lipid rafts, which are dynamic nanodomains in the plasma membrane, play a crucial role in intermembrane processes by clustering together and growing in size within the plane of the membrane while also aligning with each other across different membranes. However, the physical origin of layer by layer alignment of lipid rafts remains to be elucidated. Here, by using fluorescence imaging and synchrotron X-ray reflectivity in a phase-separated multilayer system, we find that the alignment of raft-mimicking Lo domains is regulated by the distance between bilayers. Molecular dynamics simulations reveal that the aligned state is energetically preferred when the intermembrane distance is small due to its ability to minimize the volume of surface water, which has fewer water hydrogen bonds (HBs) compared to bulk water. Our results suggest that water HB-driven alignment of lipid rafts plays a role as a precursor of intermembrane processes such as cell-cell fusion, virus entry, and signaling.


Assuntos
Ligação de Hidrogênio , Microdomínios da Membrana , Simulação de Dinâmica Molecular , Água , Água/química , Microdomínios da Membrana/química , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo
4.
EMBO J ; 39(11): e104150, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32347567

RESUMO

Alternative splicing regulates trans-synaptic adhesions and synapse development, but supporting in vivo evidence is limited. PTPδ, a receptor tyrosine phosphatase adhering to multiple synaptic adhesion molecules, is associated with various neuropsychiatric disorders; however, its in vivo functions remain unclear. Here, we show that PTPδ is mainly present at excitatory presynaptic sites by endogenous PTPδ tagging. Global PTPδ deletion in mice leads to input-specific decreases in excitatory synapse development and strength. This involves tyrosine dephosphorylation and synaptic loss of IL1RAPL1, a postsynaptic partner of PTPδ requiring the PTPδ-meA splice insert for binding. Importantly, PTPδ-mutant mice lacking the PTPδ-meA insert, and thus lacking the PTPδ interaction with IL1RAPL1 but not other postsynaptic partners, recapitulate biochemical and synaptic phenotypes of global PTPδ-mutant mice. Behaviorally, both global and meA-specific PTPδ-mutant mice display abnormal sleep behavior and non-REM rhythms. Therefore, alternative splicing in PTPδ regulates excitatory synapse development and sleep by modulating a specific trans-synaptic adhesion.


Assuntos
Proteína Acessória do Receptor de Interleucina-1/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Fases do Sono , Sinapses/metabolismo , Animais , Proteína Acessória do Receptor de Interleucina-1/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Tirosina Fosfatases/genética , Sinapses/genética
5.
Mol Psychiatry ; 27(11): 4680-4694, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35840799

RESUMO

Three-dimensional chromatin interactions regulate gene expressions. The significance of de novo mutations (DNMs) in chromatin interactions remains poorly understood for autism spectrum disorder (ASD). We generated 813 whole-genome sequences from 242 Korean simplex families to detect DNMs, and identified target genes which were putatively affected by non-coding DNMs in chromatin interactions. Non-coding DNMs in chromatin interactions were significantly involved in transcriptional dysregulations related to ASD risk. Correspondingly, target genes showed spatiotemporal expressions relevant to ASD in developing brains and enrichment in biological pathways implicated in ASD, such as histone modification. Regarding clinical features of ASD, non-coding DNMs in chromatin interactions particularly contributed to low intelligence quotient levels in ASD probands. We further validated our findings using two replication cohorts, Simons Simplex Collection (SSC) and MSSNG, and showed the consistent enrichment of non-coding DNM-disrupted chromatin interactions in ASD probands. Generating human induced pluripotent stem cells in two ASD families, we were able to demonstrate that non-coding DNMs in chromatin interactions alter the expression of target genes at the stage of early neural development. Taken together, our findings indicate that non-coding DNMs in ASD probands lead to early neurodevelopmental disruption implicated in ASD risk via chromatin interactions.


Assuntos
Transtorno do Espectro Autista , Células-Tronco Pluripotentes Induzidas , Humanos , Transtorno do Espectro Autista/genética , Cromatina/genética , Mutação/genética , Predisposição Genética para Doença/genética
6.
PLoS Biol ; 17(6): e2005326, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31166939

RESUMO

Netrin-G ligand-3 (NGL-3) is a postsynaptic adhesion molecule known to directly interact with the excitatory postsynaptic scaffolding protein postsynaptic density-95 (PSD-95) and trans-synaptically with leukocyte common antigen-related (LAR) family receptor tyrosine phosphatases to regulate presynaptic differentiation. Although NGL-3 has been implicated in the regulation of excitatory synapse development by in vitro studies, whether it regulates synapse development or function, or any other features of brain development and function, is not known. Here, we report that mice lacking NGL-3 (Ngl3-/- mice) show markedly suppressed normal brain development and postnatal survival and growth. A change of the genetic background of mice from pure to hybrid minimized these developmental effects but modestly suppressed N-methyl-D-aspartate (NMDA) receptor (NMDAR)-mediated synaptic transmission in the hippocampus without affecting synapse development, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (AMPAR)-mediated basal transmission, and presynaptic release. Intriguingly, long-term depression (LTD) was near-completely abolished in Ngl3-/- mice, and the Akt/glycogen synthase kinase 3ß (GSK3ß) signaling pathway, known to suppress LTD, was abnormally enhanced. In addition, pharmacological inhibition of Akt, but not activation of NMDARs, normalized the suppressed LTD in Ngl3-/- mice, suggesting that Akt hyperactivity suppresses LTD. Ngl3-/- mice displayed several behavioral abnormalities, including hyperactivity, anxiolytic-like behavior, impaired spatial memory, and enhanced seizure susceptibility. Among them, the hyperactivity was rapidly improved by pharmacological NMDAR activation. These results suggest that NGL-3 regulates brain development, Akt/GSK3ß signaling, LTD, and locomotive and cognitive behaviors.


Assuntos
Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Proteínas Ligadas por GPI/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Encéfalo/metabolismo , Proteínas Ligadas por GPI/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/metabolismo , Ligantes , Depressão Sináptica de Longo Prazo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Netrinas/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Sinapses/fisiologia , Transmissão Sináptica
7.
J Neurosci ; 39(10): 1910-1929, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30626696

RESUMO

The intracellular tyrosine kinase Pyk2 (PTK2B) is related to focal adhesion kinase and localizes to postsynaptic sites in brain. Pyk2 genetic variation contributes to late onset Alzheimer's disease (AD) risk. We recently observed that Pyk2 is required for synapse loss and for learning deficits in a transgenic mouse model of AD. Here, we explore the cellular and biochemical basis for the action of Pyk2 tyrosine kinase in amyloid-ß oligomer (Aßo)-induced dendritic spine loss. Overexpression of Pyk2 reduces dendritic spine density of hippocampal neurons by a kinase-dependent mechanism. Biochemical isolation of Pyk2-interacting proteins from brain identifies Graf1c, a RhoA GTPase-activating protein inhibited by Pyk2. Aßo-induced reductions in dendritic spine motility and chronic spine loss require both Pyk2 kinase and RhoA activation. Thus, Pyk2 functions at postsynaptic sites to modulate F-actin control by RhoA and regulate synapse maintenance of relevance to AD risk.SIGNIFICANCE STATEMENT Genetic variation at the Pyk2 locus is a risk for Alzheimer's disease. We have observed that Pyk2 is required for AD transgenic synapse loss and memory dysfunction. However, the cellular and biochemical basis for Pyk2 function related to AD is not defined. Here, we show that brain Pyk2 interacts with the RhoGAP protein Graf1 to alter dendritic spine stability via RhoA GTPase. Amyloid-ß oligomer-induced dendritic spine loss requires the Pyk2/Graf1 pathway.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Espinhas Dendríticas/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Hipocampo/metabolismo , Sinapses/metabolismo , Sinapses/patologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Feminino , Quinase 2 de Adesão Focal/genética , Células HEK293 , Hipocampo/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
8.
J Neurosci ; 39(4): 758-772, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30518596

RESUMO

Dozens of genes have been implicated in late onset Alzheimer's disease (AD) risk, but none has a defined mechanism of action in neurons. Here, we show that the risk factor Pyk2 (PTK2B) localizes specifically to neurons in adult brain. Absence of Pyk2 has no major effect on synapse formation or the basal parameters of synaptic transmission in the hippocampal Schaffer collateral pathway. However, the induction of synaptic LTD is suppressed in Pyk2-null slices. In contrast, deletion of Pyk2 expression does not alter LTP under control conditions. Of relevance for AD pathophysiology, Pyk2-/- slices are protected from amyloid-ß-oligomer (Aßo)-induced suppression of LTP in hippocampal slices. Acutely, a Pyk2 kinase inhibitor also prevents Aßo-induced suppression of LTP in WT slices. Female and male transgenic AD model mice expressing APPswe/PSEN1ΔE9 require Pyk2 for age-dependent loss of synaptic markers and for impairment of learning and memory. However, absence of Pyk2 does not alter Aß accumulation or gliosis. Therefore, the Pyk2 risk gene is directly implicated in a neuronal Aßo signaling pathway impairing synaptic anatomy and function.SIGNIFICANCE STATEMENT Genetic variation at the Pyk2 (PTK2B) locus is a risk for late onset Alzheimer's disease (AD), but the pathophysiological role of Pyk2 is not clear. Here, we studied Pyk2 neuronal function in mice lacking expression with and without transgenes generating amyloid-ß (Aß) plaque pathology. Pyk2 is not required for basal synaptic transmission or LTP, but participates in LTD. Hippocampal slices lacking Pyk2 are protected from AD-related Aß oligomer suppression of synaptic plasticity. In transgenic AD model mice, deletion of Pyk2 rescues synaptic loss and learning/memory deficits. Therefore, Pyk2 plays a central role in AD-related synaptic dysfunction mediating Aß-triggered dysfunction.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Quinase 2 de Adesão Focal/genética , Sinapses/patologia , Animais , Comportamento Animal , Feminino , Gliose/genética , Gliose/patologia , Aprendizagem/fisiologia , Potenciação de Longa Duração/genética , Depressão Sináptica de Longo Prazo/genética , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fatores de Risco , Transdução de Sinais/genética
9.
J Biol Chem ; 294(15): 6042-6053, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30787106

RESUMO

Oligomeric assemblies of amyloid-ß (Aß) peptide (Aßo) in the brains of individuals with Alzheimer's disease (AD) are toxic to neuronal synapses. More than a dozen Aß receptor candidates have been suggested to be responsible for various aspects of the molecular pathology and memory impairment in mouse models of AD. A lack of consistent experimental design among previous studies of different receptor candidates limits evaluation of the relative roles of these candidates, producing some controversy within the field. Here, using cell-based assays with several Aß species, including Aßo from AD brains obtained by autopsy, we directly compared the Aß-binding capacity of multiple receptor candidates while accounting for variation in expression and confirming cell surface expression. In a survey of 15 reported Aß receptors, only cellular prion protein (PrPC), Nogo receptor 1 (NgR1), and leukocyte immunoglobulin-like receptor subfamily B member 2 (LilrB2) exhibited direct binding to synaptotoxic assemblies of synthetic Aß. Both PrPC and NgR1 preferentially bound synaptotoxic oligomers rather than nontoxic monomers, and the method of oligomer preparation did not significantly alter our binding results. Hippocampal neurons lacking both NgR1 and LilrB2 exhibited a partial reduction of Aßo binding, but this reduction was lower than in neurons lacking PrPC under the same conditions. Finally, binding studies with soluble Aßo from human AD brains revealed a strong affinity for PrPC, weak affinity for NgR1, and no detectable affinity for LilrB2. These findings clarify the relative contributions of previously reported Aß receptors under controlled conditions and highlight the prominence of PrPC as an Aß-binding site.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptor Nogo 1/metabolismo , Proteínas PrPC/metabolismo , Receptores Imunológicos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Receptor Nogo 1/genética , Proteínas PrPC/genética , Receptores Imunológicos/genética
10.
Biochem Biophys Res Commun ; 501(2): 329-335, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29505793

RESUMO

Celecoxib is a non-steroidal anti-inflammatory drug that selectively inhibits cyclooxygenase-2 and is prescribed for severe pain and inflammation. The excellent therapeutic effects of celecoxib mean that it is frequently used clinically, including for women of child-bearing age. However, the prenatal effects of this compound have not been studied extensively in vertebrates. The present study examined the developmental toxicity of celecoxib using a frog embryo teratogenic assay-Xenopus (FETAX). In addition, we examined its effects on cell migration using co-cultures of human umbilical vein endothelial cells and 10T1/2 cells. These studies revealed that celecoxib induced concentration-dependent mortality and various malformations of the Xenopus internal organs, including gut miscoiling, haemorrhage, and oedema. Celecoxib also downregulated the expression of vascular wall markers (Msr and alpha smooth muscle actin) and other organ-specific markers (Nkx2.5, Cyl104 and IFABP). In vitro co-culture studies revealed that celecoxib inhibited pericyte migration and differentiation into vascular smooth muscle cells. In conclusion, celecoxib was both toxic and teratogenic in Xenopus embryos, where it produced serious heart and vessel malformation by inhibiting vascular wall maturation and vascular network formation.


Assuntos
Celecoxib/toxicidade , Teratogênicos/toxicidade , Xenopus laevis/embriologia , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Biomarcadores , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/embriologia , Celecoxib/administração & dosagem , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião não Mamífero/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Xenopus laevis/fisiologia
11.
Langmuir ; 30(48): 14369-74, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422050

RESUMO

We present a novel technique to measure diffusion coefficients of insoluble surfactant monolayers. We merge a surfactant-coated droplet with a fluorescently labeled planar monolayer. During the merging process, a monolayer on a droplet displaces the existing planar monolayer, leaving a dark area when viewed under a fluorescence microscope. We measure fractional intensities as the dyes recover, which allows diffusion coefficients to be computed. We validate this technique with the two most common phospholipid monolayers (DPPC and DOPC) and study the diffusion of their mixtures. The proposed technique has several advantages over the FRAP technique and is potentially capable of measuring the diffusion of any soluble/insoluble surfactant monolayers.


Assuntos
Fosfolipídeos/química , Tensoativos/química , 1,2-Dipalmitoilfosfatidilcolina/química , Microscopia de Fluorescência , Fosfatidilcolinas/química
12.
Cerebrovasc Dis ; 38(4): 290-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25412772

RESUMO

BACKGROUND: Intracranial arterial stenosis (ICAS) is a major cause of ischemic stroke in Asians. Despite the clinical importance of ICAS, the literature on the natural history of ICAS has been less enlightening. The aims of our study were to evaluate a long-term natural course of symptomatic and asymptomatic ICAS. METHODS: 102 subjects (37 symptomatic and 65 asymptomatic) underwent follow-up MR angiography (MRA) with a median time interval between initial and follow-up MRA of 5.7 years (range 3.6-8.5 years). For each patient, the extent of stenosis of five arteries (both middle cerebral arteries, both intracranial internal carotid arteries, and basilar artery) was classified according to five grades, by consensus: normal, mild (signal reduction <50%), moderate (signal reduction ≥50%), severe (focal signal loss with the presence of a distal signal), and occlusion. Because the sample size was too small to adjust for multiple confounders, we applied the propensity score. RESULTS: Mean (Standard deviation) age at initial MRA was 63.5 (9.6) and 54% were men. The progression rate of ICAS differed significantly between symptomatic and asymptomatic patients (22 vs. 8%, p < 0.01), indicating a 3-fold risk of progression for symptomatic stenosis compared with asymptomatic stenosis [odds ratio (OR) 3.27, 95% confidence interval (CI) 1.08-9.95]. After adjustment for propensity score, the OR was 4.84 (95% CI, 1.40-16.7). In the matched cohort, the relative risk of stenosis progression was 5.20 for symptomatic stenosis (95% CI 1.00-27.23) compared with asymptomatic stenosis. CONCLUSION: We found a greater risk of progression for symptomatic stenosis compared with asymptomatic stenosis.


Assuntos
Doenças Arteriais Cerebrais/diagnóstico , Artérias Cerebrais/patologia , Angiografia por Ressonância Magnética , Idoso , Doenças Assintomáticas , Doenças Arteriais Cerebrais/complicações , Doenças Arteriais Cerebrais/patologia , Constrição Patológica , Progressão da Doença , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Valor Preditivo dos Testes , Pontuação de Propensão , Estudos Retrospectivos , Fatores de Risco , Índice de Gravidade de Doença , Fatores de Tempo
13.
J Nanosci Nanotechnol ; 14(5): 3880-3, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24734656

RESUMO

In this work, cadmium sulfide (CdS) films were deposited on flexible polymer substrates such as polycarbonate (PC) and polyethylene terephthalate (PET). The r.f. magnetron sputtering, which is cost-effective scalable technique, was used for the film deposition. The structural and optical properties of the films grown at different sputtering pressures were investigated. When the CdS film was deposited at lower pressure, the crystallinity and the preferred orientation toward c-axis in hexagonal phase was improved. However, the optical transmittance was reduced as the sputtering pressure was decreased. Compared with the glass substrate, CdS films grown on polymer substrates were exhibited some wore structural and optical characteristics. CdTe thin film solar cell applied to sputtered CdS as a window layer showed a maximum efficiency of 11.6%.

14.
Genome Med ; 16(1): 20, 2024 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-38297291

RESUMO

BACKGROUND: Recent studies using single-cell transcriptomic analysis have reported several distinct clusters of neoplastic epithelial cells and cancer-associated fibroblasts in the pancreatic cancer tumor microenvironment. However, their molecular characteristics and biological significance have not been clearly elucidated due to intra- and inter-tumoral heterogeneity. METHODS: We performed single-cell RNA sequencing using enriched non-immune cell populations from 17 pancreatic tumor tissues (16 pancreatic cancer and one high-grade dysplasia) and generated paired spatial transcriptomic data from seven patient samples. RESULTS: We identified five distinct functional subclusters of pancreatic cancer cells and six distinct cancer-associated fibroblast subclusters. We deeply profiled their characteristics, and we found that these subclusters successfully deconvoluted most of the features suggested in bulk transcriptome analysis of pancreatic cancer. Among those subclusters, we identified a novel cancer cell subcluster, Ep_VGLL1, showing intermediate characteristics between the extremities of basal-like and classical dichotomy, despite its prognostic value. Molecular features of Ep_VGLL1 suggest its transitional properties between basal-like and classical subtypes, which is supported by spatial transcriptomic data. CONCLUSIONS: This integrative analysis not only provides a comprehensive landscape of pancreatic cancer and fibroblast population, but also suggests a novel insight to the dynamic states of pancreatic cancer cells and unveils potential therapeutic targets.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Transcriptoma , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Perfilação da Expressão Gênica , Prognóstico , Microambiente Tumoral/genética , Análise de Célula Única , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
15.
Nat Commun ; 14(1): 3547, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37321992

RESUMO

Autism spectrum disorders (ASD) represent neurodevelopmental disorders characterized by social deficits, repetitive behaviors, and various comorbidities, including epilepsy. ANK2, which encodes a neuronal scaffolding protein, is frequently mutated in ASD, but its in vivo functions and disease-related mechanisms are largely unknown. Here, we report that mice with Ank2 knockout restricted to cortical and hippocampal excitatory neurons (Ank2-cKO mice) show ASD-related behavioral abnormalities and juvenile seizure-related death. Ank2-cKO cortical neurons show abnormally increased excitability and firing rate. These changes accompanied decreases in the total level and function of the Kv7.2/KCNQ2 and Kv7.3/KCNQ3 potassium channels and the density of these channels in the enlengthened axon initial segment. Importantly, the Kv7 agonist, retigabine, rescued neuronal excitability, juvenile seizure-related death, and hyperactivity in Ank2-cKO mice. These results suggest that Ank2 regulates neuronal excitability by regulating the length of and Kv7 density in the AIS and that Kv7 channelopathy is involved in Ank2-related brain dysfunctions.


Assuntos
Epilepsia , Canais de Potássio KCNQ , Animais , Camundongos , Epilepsia/metabolismo , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Neurônios/metabolismo , Convulsões/genética , Convulsões/metabolismo
16.
J Mol Cell Cardiol ; 52(3): 718-26, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22197309

RESUMO

Despite its significant clinical implications, physiological hypertrophy remains poorly understood. In this study, the transcription coactivator Eya2 was shown to be up-regulated during physiological hypertrophy. Transgene- or adenovirus-mediated overexpression of Eya2 led to up-regulation of mTOR, a critical mediator of physiological hypertrophy. Luciferase reporter and chromatin immunoprecipitation assays revealed that Eya2 directly binds to and activates mTOR expression. The phosphorylation of mTOR downstream molecules was significantly enhanced in Eya2 transgenic (TG) hearts, implying that the Eya2-mediated induction of mTOR expression leads to an elevated mTOR activity. The transcription factor Six1 was also up-regulated during physiological hypertrophy and formed a complex with Eya2. Luciferase reporter and electrophoretic mobility shift assays revealed that the Eya2-Six1 complex binds to and enhances the expression of mTOR in a synergistic manner. Under pressure overload, Eya2 transgenic hearts developed hypertrophy which exhibited important molecular signatures of physiological hypertrophy, as assessed by gene expression profiling and measurements of expression levels of physiological hypertrophy-related genes by quantitative (q) RT-PCR. Examination of heart sections under electron microscopy revealed that the mitochondrial integrity remained largely intact in Eya2 transgenic mice, but not in wild-type littermates, under pressure overload. This finding was confirmed by measurements of mitochondrial DNA contents and the expression levels of mitochondrial function-related genes by qRT-PCR. These data suggest that Eya2 in a physical complex with Six1 plays a critical role in physiological hypertrophy. The cardioprotective effect of Eya2 appears to be due, at least in part, to its preservation of mitochondrial integrity upon pressure overload.


Assuntos
Crescimento Celular , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/ultraestrutura , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
17.
J Cell Sci ; 123(Pt 10): 1742-50, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20427313

RESUMO

SNX18 and SNX9 are members of a subfamily of SNX (sorting nexin) proteins with the same domain structure. Although a recent report showed that SNX18 and SNX9 localize differently in cells and appear to function in different trafficking pathways, concrete evidence regarding whether they act together or separately in intracellular trafficking is still lacking. Here, we show that SNX18 has a similar role to SNX9 in endocytic trafficking at the plasma membrane, rather than having a distinct role. SNX18 and SNX9 are expressed together in most cell lines, but to a different extent. Like SNX9, SNX18 interacts with dynamin and stimulates the basal GTPase activity of dynamin. It also interacts with neuronal Wiskott-Aldrich syndrome protein (N-WASP) and synaptojanin, as does SNX9. SNX18 and SNX9 can form a heterodimer and colocalize in tubular membrane structures. Depletion of SNX18 by small hairpin RNA inhibited transferrin uptake. SNX18 successfully compensates for SNX9 deficiency during clathrin-mediated endocytosis and vice versa. Total internal reflection fluorescence microscopy in living cells shows that a transient burst of SNX18 recruitment to clathrin-coated pits coincides spatiotemporally with a burst of dynamin and SNX9. Taken together, our results suggest that SNX18 functions with SNX9 in multiple pathways of endocytosis at the plasma membrane and that they are functionally redundant.


Assuntos
Membrana Celular/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Glioma/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Linhagem Celular Tumoral , Dinaminas/metabolismo , Endocitose/genética , Glioma/genética , Glioma/patologia , Humanos , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Ligação Proteica , Transporte Proteico , RNA Interferente Pequeno/genética , Nexinas de Classificação , Proteínas de Transporte Vesicular/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
18.
Aust N Z J Obstet Gynaecol ; 52(4): 348-55, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22541027

RESUMO

AIMS: The objective of this study was to derive a customised birth weight standard curve in our institute and to compare the perinatal outcomes of small-for-gestational-age (SGA) births classified by population-based versus customised birth weight standards. METHODS: We surveyed 9052 normal singleton deliveries and generated customised standards by adjusting for maternal characteristics and neonatal gender. We compared adverse perinatal outcomes between SGA and non-SGA births classified by both standards. RESULTS: According to the population-based standards, mothers of SGA infants were younger, thinner and shorter and had higher rates of nulliparity and female births. We adjusted for these maternal characteristics and neonatal gender in our customised standards. Multivariate analysis revealed that there were no differences in neonatal composite morbidity between the standards. However, infants classified as SGA by the customised standards showed a significantly higher rate of neonatal intensive care unit (NICU) admission than those classified by the population-based standards. CONCLUSION: Our study showed that customised SGA made no significant differences in neonatal composite morbidity, only a modest increase in NICU admission rate compared to population-based standard. To clarify the association of adverse perinatal outcomes with customised SGA, larger studies are required.


Assuntos
Peso ao Nascer , Recém-Nascido Pequeno para a Idade Gestacional , Mães/estatística & dados numéricos , Adulto , Análise de Variância , Efeito de Coortes , Feminino , Humanos , Recém-Nascido , Masculino , Idade Materna , Análise Multivariada , Padrões de Referência , Valores de Referência , República da Coreia , Estudos Retrospectivos , Estatísticas não Paramétricas
19.
J Cell Biol ; 221(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35819332

RESUMO

IRSp53 (aka BAIAP2) is a scaffold protein that couples membranes with the cytoskeleton in actin-filled protrusions such as filopodia and lamellipodia. The protein is abundantly expressed in excitatory synapses and is essential for synapse development and synaptic plasticity, although with poorly understood mechanisms. Here we show that specific multivalent interactions between IRSp53 and its binding partners PSD-95 or Shank3 drive phase separation of the complexes in solution. IRSp53 can be enriched to the reconstituted excitatory PSD (ePSD) condensates via bridging to the core and deeper layers of ePSD. Overexpression of a mutant defective in the IRSp53/PSD-95 interaction perturbs synaptic enrichment of IRSp53 in mouse cortical neurons. The reconstituted PSD condensates promote bundled actin filament formation both in solution and on membranes, via IRSp53-mediated actin binding and bundling. Overexpression of mutants that perturb IRSp53-actin interaction leads to defects in synaptic maturation of cortical neurons. Together, our studies provide potential mechanistic insights into the physiological roles of IRSp53 in synapse formation and function.


Assuntos
Actinas , Proteínas do Tecido Nervoso , Densidade Pós-Sináptica , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Densidade Pós-Sináptica/metabolismo , Pseudópodes/genética , Pseudópodes/metabolismo , Sinapses/genética , Sinapses/metabolismo
20.
Mol Neurodegener ; 17(1): 32, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35501917

RESUMO

BACKGROUND: Genetic variation at the PTK2B locus encoding the protein Pyk2 influences Alzheimer's disease risk. Neurons express Pyk2 and the protein is required for Amyloid-ß (Aß) peptide driven deficits of synaptic function and memory in mouse models, but Pyk2 deletion has minimal effect on neuro-inflammation. Previous in vitro data suggested that Pyk2 activity might enhance GSK3ß-dependent Tau phosphorylation and be required for tauopathy. Here, we examine the influence of Pyk2 on Tau phosphorylation and associated pathology. METHODS: The effect of Pyk2 on Tau phosphorylation was examined in cultured Hek cells through protein over-expression and in iPSC-derived human neurons through pharmacological Pyk2 inhibition. PS19 mice overexpressing the P301S mutant of human Tau were employed as an in vivo model of tauopathy. Phenotypes of PS19 mice with a targeted deletion of Pyk2 expression were compared with PS19 mice with intact Pyk2 expression. Phenotypes examined included Tau phosphorylation, Tau accumulation, synapse loss, gliosis, proteomic profiling and behavior. RESULTS: Over-expression experiments from Hek293T cells indicated that Pyk2 contributed to Tau phosphorylation, while iPSC-derived human neuronal cultures with endogenous protein levels supported the opposite conclusion. In vivo, multiple phenotypes of PS19 were exacerbated by Pyk2 deletion. In Pyk2-null PS19 mice, Tau phosphorylation and accumulation increased, mouse survival decreased, spatial memory was impaired and hippocampal C1q deposition increased relative to PS19 littermate controls. Proteomic profiles of Pyk2-null mouse brain revealed that several protein kinases known to interact with Tau are regulated by Pyk2. Endogenous Pyk2 suppresses LKB1 and p38 MAPK activity, validating one potential pathway contributing to increased Tau pathology. CONCLUSIONS: The absence of Pyk2 results in greater mutant Tau-dependent phenotypes in PS19 mice, in part via increased LKB1 and MAPK activity. These data suggest that in AD, while Pyk2 activity mediates Aß-driven deficits, Pyk2 suppresses Tau-related phenotypes.


Assuntos
Doença de Alzheimer , Tauopatias , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Quinase 2 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Fosforilação , Proteômica , Tauopatias/metabolismo , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA