Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Biochem Cell Biol ; 98(2): 299-306, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31965814

RESUMO

Ankyrin-B (AnkB) is scaffolding protein that anchors integral membrane proteins to the cardiomyocyte cytoskeleton. We recently identified an AnkB variant, AnkB p.S646F (ANK2 c.1937 C>T) associated with a phenotype ranging from predisposition for cardiac arrhythmia to cardiomyopathy. AnkB p.S646F exhibited reduced expression levels in the H9c2 rat ventricular-derived cardiomyoblast cell line relative to wildtype AnkB. Here, we demonstrate that AnkB is regulated by proteasomal degradation and proteasome inhibition rescues AnkB p.S646F expression levels in H9c2 cells, although this effect is not conserved with differentiation. We also compared the impact of wildtype AnkB and AnkB p.S646F on cell viability and proliferation. AnkB p.S646F expression resulted in decreased cell viability at 30 h after transfection, whereas we observed a greater proportion of cycling, Ki67-positive cells at 48 h after transfection. Notably, the number of GFP-positive cells was low and was consistent between wildtype AnkB and AnkB p.S646F expressing cells, suggesting that AnkB and AnkB p.S646F affected paracrine communication between H9c2 cells differentially. This work reveals that AnkB levels are regulated by the proteasome and that AnkB p.S646F compromises cell viability. Together, these findings provide key new insights into the putative cellular and molecular mechanisms of AnkB-related cardiac disease.


Assuntos
Anquirinas/metabolismo , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Cardiomiopatias , Comunicação Celular , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Citoesqueleto/metabolismo , Imuno-Histoquímica , Microscopia Confocal , Fenótipo , Ratos
2.
J Neurosci ; 36(4): 1203-10, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26818508

RESUMO

We demonstrated previously that Pannexin 1 (Panx1), an ion and metabolite channel, promotes the growth and proliferation of ventricular zone (VZ) neural precursor cells (NPCs) in vitro. To investigate its role in vivo, we used floxed Panx1 mice in combination with viruses to delete Panx1 in VZ NPCs and to track numbers of Panx1-null and Panx1-expressing VZ NPCs over time. Two days after virus injection, Panx1-null cells were less abundant than Panx1-expressing cells, suggesting that Panx1 is required for the maintenance of VZ NPCs. We also investigated the effect of Panx1 deletion in VZ NPCs after focal cortical stroke via photothrombosis. Panx1 is essential for maintaining elevated VZ NPC numbers after stroke. In contrast, Panx1-null NPCs were more abundant than Panx1-expressing NPCs in the peri-infarct cortex. Together, these findings suggest that Panx1 plays an important role in NPC maintenance in the VZ niche in the naive and stroke brain and could be a key target for improving NPC survival in the peri-infarct cortex. SIGNIFICANCE STATEMENT: Here, we demonstrate that Pannexin 1 (Panx1) maintains a consistent population size of neural precursor cells in the ventricular zone, both in the healthy brain and in the context of stroke. In contrast, Panx1 appears to be detrimental to the survival of neural precursor cells that surround damaged cortical tissue in the stroke brain. This suggests that targeting Panx1 in the peri-infarct cortex, in combination with other therapies, could improve cell survival around the injury site.


Assuntos
Infarto Cerebral/patologia , Ventrículos Cerebrais/citologia , Conexinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Análise de Variância , Animais , Caspase 3/metabolismo , Contagem de Células , Sobrevivência Celular/fisiologia , Conexinas/genética , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neuropeptídeos/metabolismo , Acidente Vascular Cerebral/complicações
4.
Front Cardiovasc Med ; 9: 964675, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35990955

RESUMO

The ANK2 gene encodes for ankyrin-B (ANKB), one of 3 members of the ankyrin family of proteins, whose name is derived from the Greek word for anchor. ANKB was originally identified in the brain (B denotes "brain") but has become most widely known for its role in cardiomyocytes as a scaffolding protein for ion channels and transporters, as well as an interacting protein for structural and signaling proteins. Certain loss-of-function ANK2 variants are associated with a primarily cardiac-presenting autosomal-dominant condition with incomplete penetrance and variable expressivity characterized by a predisposition to supraventricular and ventricular arrhythmias, arrhythmogenic cardiomyopathy, congenital and adult-onset structural heart disease, and sudden death. Another independent group of ANK2 variants are associated with increased risk for distinct neurological phenotypes, including epilepsy and autism spectrum disorders. The mechanisms underlying ANKB's roles in cells in health and disease are not fully understood; however, several clues from a range of molecular and cell biological studies have emerged. Notably, ANKB exhibits several isoforms that have different cell-type-, tissue-, and developmental stage- expression profiles. Given the conservation within ankyrins across evolution, model organism studies have enabled the discovery of several ankyrin roles that could shed important light on ANKB protein-protein interactions in heart and brain cells related to the regulation of cellular polarity, organization, calcium homeostasis, and glucose and fat metabolism. Along with this accumulation of evidence suggesting a diversity of important ANKB cellular functions, there is an on-going debate on the role of ANKB in disease. We currently have limited understanding of how these cellular functions link to disease risk. To this end, this review will examine evidence for the cellular roles of ANKB and the potential contribution of ANKB functional variants to disease risk and presentation. This contribution will highlight the impact of ANKB dysfunction on cardiac and neuronal cells and the significance of understanding the role of ANKB variants in disease.

5.
Front Neuroanat ; 15: 722443, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34949993

RESUMO

The ever-expanding availability and evolution of microscopy tools has enabled ground-breaking discoveries in neurobiology, particularly with respect to the analysis of cell-type density and distribution. Widespread implementation of many of the elegant image processing tools available continues to be impeded by the lack of complete workflows that span from experimental design, labeling techniques, and analysis workflows, to statistical methods and data presentation. Additionally, it is important to consider open science principles (e.g., open-source software and tools, user-friendliness, simplicity, and accessibility). In the present methodological article, we provide a compendium of resources and a FIJI-ImageJ-based workflow aimed at improving the quantification of cell density in mouse brain samples using semi-automated open-science-based methods. Our proposed framework spans from principles and best practices of experimental design, histological and immunofluorescence staining, and microscopy imaging to recommendations for statistical analysis and data presentation. To validate our approach, we quantified neuronal density in the mouse barrel cortex using antibodies against pan-neuronal and interneuron markers. This framework is intended to be simple and yet flexible, such that it can be adapted to suit distinct project needs. The guidelines, tips, and proposed methodology outlined here, will support researchers of wide-ranging experience levels and areas of focus in neuroscience research.

6.
Pharmacol Ther ; 225: 107840, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33753132

RESUMO

Purinergic signaling encompasses the cycle of adenosine 5' triphosphate (ATP) release and its metabolism into nucleotide and nucleoside derivatives, the direct release of nucleosides, and subsequent receptor-triggered downstream intracellular pathways. Since the discovery of nerve terminal and glial ATP release into the neuropil, purinergic signaling has been implicated in the modulation of nervous system development, function, and disease. In this review, we detail our current understanding of the roles of the pannexin 1 (PANX1) ATP-release channel in neuronal development and plasticity, glial signaling, and neuron-glial-immune interactions. We additionally provide an overview of PANX1 structure, activation, and permeability to orientate readers and highlight recent research developments. We identify areas of convergence between PANX1 and purinergic receptor actions. Additional highlights include data on PANX1's participation in the pathophysiology of nervous system developmental, degenerative, and inflammatory disorders. Our aim in combining this knowledge is to facilitate the movement of our current understanding of PANX1 in the context of other nervous system purinergic signaling mechanisms one step closer to clinical translation.


Assuntos
Trifosfato de Adenosina , Conexinas , Proteínas do Tecido Nervoso , Transdução de Sinais , Trifosfato de Adenosina/metabolismo , Conexinas/metabolismo , Humanos , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso , Neurônios
7.
eNeuro ; 7(4)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32737184

RESUMO

The integration of neurons into networks relies on the formation of dendritic spines. These specialized structures arise from dynamic filopodia-like dendritic protrusions. It was recently reported that cortical neurons lacking the channel protein pannexin 1 (PANX1) exhibited higher dendritic spine densities. Here, we expanded on those findings to investigate, at an earlier developmental time point (with more abundant dendritic protrusions), whether differences in the properties of dendritic protrusion dynamics could contribute to this previously discovered phenomenon. Using a fluorescent membrane tag (mCherry-CD9-10) to visualize dendritic protrusions in developing neurons [at 10 d in vitro (DIV10)], we confirmed that lack of PANX1 led to higher protrusion density, while transient transfection of Panx1 led to decreased protrusion density. To quantify the impact of PANX1 expression on protrusion formation, elimination, and motility, we used live cell imaging in DIV10 neurons (one frame every 5 s for 10 min). We discovered that at DIV10, loss of PANX1 stabilized protrusions. Notably, re-expression of PANX1 in Panx1 knock-out (KO) neurons resulted in a significant increase in protrusion motility and turnover. In summary, these new data revealed that PANX1 could regulate the development of dendritic spines, in part, by controlling dendritic protrusion dynamics.


Assuntos
Espinhas Dendríticas , Células-Tronco Neurais , Neurônios , Pseudópodes
8.
Cell Calcium ; 90: 102253, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32688074

RESUMO

A new study by Yang and colleagues has revealed that TNF-alpha regulates PANX1 levels through an NF-kB-dependent mechanism in human endothelial cells. PANX1 modulates Ca2+ influx contributing to IL-1beta production independent of purinergic signaling. These novel findings expand our understanding of TNF-alpha-mediated upregulation of IL-1beta with implications for responses to tissue injury and infection.


Assuntos
Conexinas/metabolismo , Infecções/metabolismo , Infecções/patologia , Inflamação/metabolismo , Inflamação/patologia , Proteínas do Tecido Nervoso/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamassomos/metabolismo , Modelos Biológicos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
9.
Mol Brain ; 12(1): 75, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31477143

RESUMO

Ankyrin B (AnkB) is an adaptor and scaffold for motor proteins and various ion channels that is ubiquitously expressed, including in the brain. AnkB has been associated with neurological disorders such as epilepsy and autism spectrum disorder, but understanding of the underlying mechanisms is limited. Cav2.1, the pore-forming subunit of P/Q type voltage gated calcium channels, is a known interactor of AnkB and plays a crucial role in neuronal function. Here we report that wildtype AnkB increased overall Cav2.1 levels without impacting surface Cav2.1 levels in HEK293T cells. An AnkB variant, p.S646F, which we recently discovered to be associated with seizures, further increased overall Cav2.1 levels, again with no impact on surface Cav2.1 levels. AnkB p.Q879R, on the other hand, increased surface Cav2.1 levels in the presence of accessory subunits α2δ1 and ß4. Additionally, AnkB p.E1458G decreased surface Cav2.1 irrespective of the presence of accessory subunits. In addition, we found that partial deletion of AnkB in cortex resulted in a decrease in overall Cav2.1 levels, with no change to the levels of Cav2.1 detected in synaptosome fractions. Our work suggests that depending on the particular variant, AnkB regulates intracellular and surface Cav2.1. Notably, expression of the AnkB variant associated with seizure (AnkB p.S646F) caused further increase in intracellular Cav2.1 levels above that of even wildtype AnkB. These novel findings have important implications for understanding the role of AnkB and Cav2.1 in the regulation of neuronal function in health and disease.


Assuntos
Anquirinas/metabolismo , Canais de Cálcio Tipo N/metabolismo , Membrana Celular/metabolismo , Espaço Intracelular/metabolismo , Proteínas Mutantes/metabolismo , Animais , Anquirinas/genética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação/genética , Subunidades Proteicas/metabolismo , Sinapses/metabolismo
10.
eNeuro ; 6(3)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118206

RESUMO

Dendritic spines are the postsynaptic targets of excitatory synaptic inputs that undergo extensive proliferation and maturation during the first postnatal month in mice. However, our understanding of the molecular mechanisms that regulate spines during this critical period is limited. Previous work has shown that pannexin 1 (Panx1) regulates neurite growth and synaptic plasticity. We therefore investigated the impact of global Panx1 KO on spontaneous cortical neuron activity using Ca2+ imaging and in silico network analysis. Panx1 KO increased both the number and size of spontaneous co-active cortical neuron network ensembles. To understand the basis for these findings, we investigated Panx1 expression in postnatal synaptosome preparations from early postnatal mouse cortex. Between 2 and 4 postnatal weeks, we observed a precipitous drop in cortical synaptosome protein levels of Panx1, suggesting it regulates synapse proliferation and/or maturation. At the same time points, we observed significant enrichment of the excitatory postsynaptic density proteins PSD-95, GluA1, and GluN2a in cortical synaptosomes from global Panx1 knock-out mice. Ex vivo analysis of pyramidal neuron structure in somatosensory cortex revealed a consistent increase in dendritic spine densities in both male and female Panx1 KO mice. Similar findings were observed in an excitatory neuron-specific Panx1 KO line (Emx1-Cre driven; Panx1 cKOE) and in primary Panx1 KO cortical neurons cultured in vitro. Altogether, our study suggests that Panx1 negatively regulates cortical dendritic spine development.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Conexinas/fisiologia , Espinhas Dendríticas/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Sinalização do Cálcio , Córtex Cerebral/metabolismo , Conexinas/genética , Conexinas/metabolismo , Espinhas Dendríticas/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Imagem Óptica , Sinaptossomos/metabolismo , Sinaptossomos/fisiologia
11.
Sci Rep ; 9(1): 9721, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31278290

RESUMO

The Pannexin 1 (Panx1) ion and metabolite channel is expressed in a wide variety of cells where it regulates a number of cell behaviours including proliferation and differentiation. Panx1 is expressed on the cell surface as well as intracellular membranes. Previous work suggests that a region within the proximal Panx1 C-terminus (Panx1CT) regulates cell surface localization. Here we report the discovery of a putative leucine-rich repeat (LRR) motif in the proximal Panx1CT necessary for Panx1 cell surface expression in HEK293T cells. Deletion of the putative LRR motif results in significant loss of Panx1 cell surface distribution. Outcomes of complementary cell surface oligomerization and glycosylation state analyses were consistent with reduced cell surface expression of Panx1 LRR deletion mutants. Of note, the oligomerization analysis revealed the presence of putative dimers and trimers of Panx1 at the cell surface. Expression of Panx1 increased HEK293T cell growth and reduced doubling time, while expression of a Panx1 LRR deletion mutant (highly conserved segment) did not reproduce this effect. In summary, here we discovered the presence of a putative LRR motif in the Panx1CT that impacts on Panx1 cell surface localization. Overall these findings provide new insights into the molecular mechanisms underlying C-terminal regulation of Panx1 trafficking and raise potential new lines of investigation with respect to Panx1 oligomerization and glycosylation.


Assuntos
Membrana Celular/metabolismo , Conexinas/química , Conexinas/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Motivos de Aminoácidos , Conexinas/genética , Glicosilação , Células HEK293 , Humanos , Proteínas do Tecido Nervoso/genética , Multimerização Proteica , Transporte Proteico , Deleção de Sequência
12.
Front Cell Neurosci ; 12: 124, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867357

RESUMO

Neurite formation relies on finely-tuned control of the cytoskeleton. Here we identified a novel protein-protein interaction between the ion and metabolite channel protein Pannexin 1 (Panx1) and collapsin response mediator protein 2 (Crmp2), a positive regulator of microtubule polymerization and stabilization. Panx1 and Crmp2 co-precipitated from both Neuro-2a (N2a) cells and mouse ventricular zone (VZ) tissue. In vitro binding assays between purified proteins revealed the interaction occurs directly between the Panx1 C-terminus (Panx1 CT) and Crmp2. Because Crmp2 is a well-established microtubule-stabilizing protein, and we previously observed a marked increase in neurite formation following treatment with the Panx1 blocker, probenecid, in N2a cells and VZ neural precursor cells (NPCs), we investigated the impact of probenecid on the Panx1-Crmp2 interaction. Probenecid treatment significantly disrupted the Panx1-Crmp2 interaction by both immunoprecipitation (IP) and proximity ligation analysis, without altering overall Crmp2 protein expression levels. In the presence of probenecid, Crmp2 was concentrated at the distal ends of growing neurites. Moreover, probenecid treatment increased tubulin polymerization and microtubule stability in N2a cells. These results reveal that probenecid disrupts a novel interaction between Panx1 and the microtubule stabilizer, Crmp2, and also increases microtubule stability.

13.
Proteomics Clin Appl ; 11(9-10)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28508575

RESUMO

PURPOSE: After cortical stroke, neural precursor cells (NPCs) in the distal ventricular zone (VZ) proliferate more rapidly and migrate toward the injured cortex. While evidence suggests this can enhance stroke recovery, the underlying molecular mechanisms initiating the response are poorly understood. Here we identified changes in protein expression in the ipsilateral VZ early (4 h) after stroke to gain insight into the initial mechanisms involved in NPC activation post-stroke. EXPERIMENTAL DESIGN: Four hours after photothrombotic stroke (or sham surgery control) in the sensorimotor cortex, adult mice (10 stroke, 10 sham) were subjected to cardiac perfusion with PBS, and ipsilateral and contralateral VZ tissue was microdissected. Two separate sets of ipsilateral and contralateral VZ tissues (from 5 pooled surgery or 5 pooled sham mice) were analyzed simultaneously using 8-plex iTRAQ. We used Western blotting and confocal microscopy to confirm changes in protein expression in the VZ ipsilateral to stroke in a separate cohort of mice. RESULTS: We identified nine proteins which exhibited a significant mean increase (by ≥ 2-fold) in stroke ipsilateral compared to sham ipsilateral. Many of these proteins were antiproteases or cytokine/growth factor binding proteins that are known to act as inflammatory responders or effectors and play roles in modulating tissue growth and remodeling. CONCLUSION AND CLINICAL RELEVANCE: These novel findings support a growing body of literature that inflammatory signaling is involved in the NPC response to brain injury and identifies novel potential targets that could be exploited to better understand and to optimize this regenerative response.


Assuntos
Mediadores da Inflamação/metabolismo , Células-Tronco Neurais/metabolismo , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Regulação para Cima , Animais , Ontologia Genética , Camundongos , Acidente Vascular Cerebral/genética
14.
Neural Regen Res ; 11(10): 1540-1544, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27904473

RESUMO

We recently reported that targeted deletion of Pannexin 1 in neural precursor cells of the ventricular zone impairs the maintenance of these cells in healthy and stroke-injured brain. Here we frame this exciting new finding in the context of our previous studies on Pannexin 1 in neural precursors as well as the close relationship between Pannexin 1 and purinergic receptors established by other groups. Moreover, we identify important gaps in our understanding of Pannexin 1 in neural precursor cell biology in terms of the underlying molecular mechanisms and functional/behavioural outcomes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA