Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS One ; 17(7): e0270915, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35776750

RESUMO

It is widely anticipated that a reduction of brain levels of the cellular prion protein (PrPC) can prolong survival in a group of neurodegenerative diseases known as prion diseases. To date, efforts to decrease steady-state PrPC levels by targeting this protein directly with small molecule drug-like compounds have largely been unsuccessful. Recently, we reported Na,K-ATPases to reside in immediate proximity to PrPC in the brain, unlocking an opportunity for an indirect PrPC targeting approach that capitalizes on the availability of potent cardiac glycosides (CGs). Here, we report that exposure of human co-cultures of neurons and astrocytes to non-toxic nanomolar levels of CGs causes profound reductions in PrPC levels. The mechanism of action underpinning this outcome relies primarily on a subset of CGs engaging the ATP1A1 isoform, one of three α subunits of Na,K-ATPases expressed in brain cells. Upon CG docking to ATP1A1, the ligand receptor complex, and PrPC along with it, is internalized by the cell. Subsequently, PrPC is channeled to the lysosomal compartment where it is digested in a manner that can be rescued by silencing the cysteine protease cathepsin B. These data signify that the repurposing of CGs may be beneficial for the treatment of prion disorders.


Assuntos
Glicosídeos Cardíacos , Doenças Priônicas , Príons , Adenosina Trifosfatases , Glicosídeos Cardíacos/farmacologia , Humanos , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Proteínas Priônicas/metabolismo , Príons/metabolismo
2.
PLoS One ; 16(11): e0258682, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34847154

RESUMO

The prion protein (PrP) is best known for its ability to cause fatal neurodegenerative diseases in humans and animals. Here, we revisited its molecular environment in the brain using a well-developed affinity-capture mass spectrometry workflow that offers robust relative quantitation. The analysis confirmed many previously reported interactions. It also pointed toward a profound enrichment of Na,K-ATPases (NKAs) in proximity to cellular PrP (PrPC). Follow-on work validated the interaction, demonstrated partial co-localization of the ATP1A1 and PrPC, and revealed that cells exposed to cardiac glycoside (CG) inhibitors of NKAs exhibit correlated changes to the steady-state levels of both proteins. Moreover, the presence of PrPC was observed to promote the ion uptake activity of NKAs in a human co-culture paradigm of differentiated neurons and glia cells, and in mouse neuroblastoma cells. Consistent with this finding, changes in the expression of 5'-nucleotidase that manifest in wild-type cells in response to CG exposure can also be observed in untreated PrPC-deficient cells. Finally, the endoproteolytic cleavage of the glial fibrillary acidic protein, a hallmark of late-stage prion disease, can also be induced by CGs, raising the prospect that a loss of NKA activity may contribute to the pathobiology of prion diseases.


Assuntos
Proteínas Priônicas/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , 5'-Nucleotidase/metabolismo , Animais , Encéfalo/metabolismo , Calpaína/metabolismo , Glicosídeos Cardíacos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Modelos Biológicos , Proteínas Priônicas/deficiência , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/metabolismo , Reprodutibilidade dos Testes
3.
J Biol Chem ; 297(3): 101073, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34390689

RESUMO

The study of prions and the discovery of candidate therapeutics for prion disease have been facilitated by the ability of prions to replicate in cultured cells. Paradigms in which prion proteins from different species are expressed in cells with low or no expression of endogenous prion protein (PrP) have expanded the range of prion strains that can be propagated. In these systems, cells stably expressing a PrP of interest are typically generated via coexpression of a selectable marker and treatment with an antibiotic. Here, we report the unexpected discovery that the aminoglycoside G418 (Geneticin) interferes with the ability of stably transfected cultured cells to become infected with prions. In G418-resistant lines of N2a or CAD5 cells, the presence of G418 reduced levels of protease-resistant PrP following challenge with the RML or 22L strains of mouse prions. G418 also interfered with the infection of cells expressing hamster PrP with the 263K strain of hamster prions. Interestingly, G418 had minimal to no effect on protease-resistant PrP levels in cells with established prion infection, arguing that G418 selectively interferes with de novo prion infection. As G418 treatment had no discernible effect on cellular PrP levels or its localization, this suggests that G418 may specifically target prion assemblies or processes involved in the earliest stages of prion infection.


Assuntos
Gentamicinas/farmacologia , Proteínas Priônicas/efeitos dos fármacos , Príons/antagonistas & inibidores , Aminoglicosídeos/metabolismo , Aminoglicosídeos/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Gentamicinas/metabolismo , Camundongos , Proteínas PrPC/efeitos dos fármacos , Proteínas PrPC/metabolismo , Proteínas PrPSc/efeitos dos fármacos , Proteínas PrPSc/metabolismo , Doenças Priônicas/prevenção & controle , Proteínas Priônicas/metabolismo , Príons/metabolismo , Inibidores da Síntese de Proteínas
4.
Biol Rev Camb Philos Soc ; 96(5): 1907-1932, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33960099

RESUMO

The quest to determine the function of a protein can represent a profound challenge. Although this task is the mandate of countless research groups, a general framework for how it can be approached is conspicuously lacking. Moreover, even expectations for when the function of a protein can be considered to be 'known' are not well defined. In this review, we begin by introducing concepts pertinent to the challenge of protein function assignments. We then propose a framework for inferring a protein's function from four data categories: 'inheritance', 'distribution', 'interactions' and 'phenotypes' (IDIP). We document that the functions of proteins emerge at the intersection of inferences drawn from these data categories and emphasise the benefit of considering them in an evolutionary context. We then apply this approach to the cellular prion protein (PrPC ), well known for its central role in prion diseases, whose function continues to be considered elusive by many investigators. We document that available data converge on the conclusion that the function of the prion protein is to control a critical post-translational modification of the neural cell adhesion molecule in the context of epithelial-to-mesenchymal transition and related plasticity programmes. Finally, we argue that this proposed function of PrPC has already passed the test of time and is concordant with the IDIP framework in a way that other functions considered for this protein fail to achieve. We anticipate that the IDIP framework and the concepts analysed herein will aid the investigation of other proteins whose primary functional assignments have thus far been intractable.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas Priônicas , Proteínas Priônicas/genética , Proteínas Priônicas/metabolismo , Processamento de Proteína Pós-Traducional
5.
PLoS One ; 14(5): e0217392, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31136617

RESUMO

Somatostatin (SST) is a cyclic peptide that is understood to inhibit the release of hormones and neurotransmitters from a variety of cells by binding to one of five canonical G protein-coupled SST receptors (SSTR1 to SSTR5). Recently, SST was also observed to interact with the amyloid beta (Aß) peptide and affect its aggregation kinetics, raising the possibility that it may bind other brain proteins. Here we report on an SST interactome analysis that made use of human brain extracts as biological source material and incorporated advanced mass spectrometry workflows for the relative quantitation of SST binding proteins. The analysis revealed SST to predominantly bind several members of the P-type family of ATPases. Subsequent validation experiments confirmed an interaction between SST and the sodium-potassium pump (Na+/K+-ATPase) and identified a tryptophan residue within SST as critical for binding. Functional analyses in three different cell lines indicated that SST might negatively modulate the K+ uptake rate of the Na+/K+-ATPase.


Assuntos
Encéfalo/metabolismo , ATPases do Tipo-P/metabolismo , Somatostatina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Linhagem Celular , Células HEK293 , Humanos , Cinética , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Radioisótopos de Rubídio/farmacocinética , ATPase Trocadora de Sódio-Potássio/metabolismo , Somatostatina-28/metabolismo
6.
J Biol Chem ; 294(13): 4911-4923, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30705093

RESUMO

Prions are infectious protein aggregates that cause several fatal neurodegenerative diseases. Prion research has been hindered by a lack of cellular paradigms for studying the replication of prions from different species. Although hamster prions have been widely used to study prion replication in animals and within in vitro amplification systems, they have proved challenging to propagate in cultured cells. Because the murine catecholaminergic cell line CAD5 is susceptible to a diverse range of mouse prion strains, we hypothesized that it might also be capable of propagating nonmouse prions. Here, using CRISPR/Cas9-mediated genome engineering, we demonstrate that CAD5 cells lacking endogenous mouse PrP expression (CAD5-PrP-/- cells) can be chronically infected with hamster prions following stable expression of hamster PrP. When exposed to the 263K, HY, or 139H hamster prion strains, these cells stably propagated high levels of protease-resistant PrP. Hamster prion replication required absence of mouse PrP, and hamster PrP inhibited the propagation of mouse prions. Cellular homogenates from 263K-infected cells exhibited prion seeding activity in the RT-QuIC assay and were infectious to naïve cells expressing hamster PrP. Interestingly, murine N2a neuroblastoma cells ablated for endogenous PrP expression were susceptible to mouse prions, but not hamster prions upon expression of cognate PrP, suggesting that CAD5 cells either possess cellular factors that enhance or lack factors that restrict the diversity of prion strains that can be propagated. We conclude that transfected CAD5-PrP-/- cells may be a useful tool for assessing the biology of prion strains and dissecting the mechanism of prion replication.


Assuntos
Príons/metabolismo , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Cricetinae , Edição de Genes , Camundongos , Príons/genética
7.
Sci Rep ; 8(1): 8654, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29872131

RESUMO

At times, it can be difficult to discern if a lack of overlap in reported interactions for a protein-of-interest reflects differences in methodology or biology. In such instances, systematic analyses of protein-protein networks across diverse paradigms can provide valuable insights. Here, we interrogated the interactome of the prion protein (PrP), best known for its central role in prion diseases, in four mouse cell lines. Analyses made use of identical affinity capture and sample processing workflows. Negative controls were generated from PrP knockout lines of the respective cell models, and the relative levels of peptides were quantified using isobaric labels. The study uncovered 26 proteins that reside in proximity to PrP. All of these proteins are predicted to have access to the outer face of the plasma membrane, and approximately half of them were not reported to interact with PrP before. Strikingly, although several proteins exhibited profound co-enrichment with PrP in a given model, except for the neural cell adhesion molecule 1, no protein was highly enriched in all PrP-specific interactomes. However, Gene Ontology analyses revealed a shared association of the majority of PrP candidate interactors with cellular events at the intersection of transforming growth factor ß and integrin signaling.


Assuntos
Integrinas/metabolismo , Proteínas Priônicas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Camundongos , Proteínas Priônicas/genética , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas
8.
Sci Rep ; 7: 40313, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-28098160

RESUMO

The prion protein (PrP) evolved from the subbranch of ZIP metal ion transporters comprising ZIPs 5, 6 and 10, raising the prospect that the study of these ZIPs may reveal insights relevant for understanding the function of PrP. Building on data which suggested PrP and ZIP6 are critical during epithelial-to-mesenchymal transition (EMT), we investigated ZIP6 in an EMT paradigm using ZIP6 knockout cells, mass spectrometry and bioinformatic methods. Reminiscent of PrP, ZIP6 levels are five-fold upregulated during EMT and the protein forms a complex with NCAM1. ZIP6 also interacts with ZIP10 and the two ZIP transporters exhibit interdependency during their expression. ZIP6 contributes to the integration of NCAM1 in focal adhesion complexes but, unlike cells lacking PrP, ZIP6 deficiency does not abolish polysialylation of NCAM1. Instead, ZIP6 mediates phosphorylation of NCAM1 on a cluster of cytosolic acceptor sites. Substrate consensus motif features and in vitro phosphorylation data point toward GSK3 as the kinase responsible, and interface mapping experiments identified histidine-rich cytoplasmic loops within the ZIP6/ZIP10 heteromer as a novel scaffold for GSK3 binding. Our data suggests that PrP and ZIP6 inherited the ability to interact with NCAM1 from their common ZIP ancestors but have since diverged to control distinct posttranslational modifications of NCAM1.


Assuntos
Antígeno CD56/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Transição Epitelial-Mesenquimal , Adesões Focais/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte de Cátions/química , Citoesqueleto/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Histidina/metabolismo , Humanos , Integrinas/metabolismo , Camundongos , Modelos Biológicos , Ácido N-Acetilneuramínico/metabolismo , Fosforilação , Proteínas Priônicas/metabolismo , Ligação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína
9.
ASN Neuro ; 8(6)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27879349

RESUMO

Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.


Assuntos
Antígeno CD56/metabolismo , Proteínas Priônicas/metabolismo , Sialiltransferases/fisiologia , Animais , Antígeno CD56/genética , Humanos , Neurogênese , Proteínas Priônicas/genética , Mapas de Interação de Proteínas , Ácidos Siálicos/metabolismo , Transdução de Sinais
10.
PLoS One ; 11(6): e0156779, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27327609

RESUMO

A popular method for studying the function of a given protein is to generate and characterize a suitable model deficient for its expression. For the prion protein (PrP), best known for its role in several invariably fatal neurodegenerative diseases, a natural choice, therefore, would be to undertake such studies with brain samples. We recently documented the surprising observation that PrP deficiency caused a loss or enhancement of NCAM1 polysialylation, dependent on the cell model used. To identify possible causes for this disparity, we set out to systematically investigate the consequence of PrP deficiency on the global proteome in brain tissue and in four distinct cell models. Here we report that PrP deficiency causes robust but surprisingly divergent changes to the global proteomes of cell models but has no discernible impact on the global brain proteome. Amongst >1,500 proteins whose levels were compared in wild-type and PrP-deficient models, members of the MARCKS protein family exhibited pronounced, yet cell model-dependent changes to their steady-state levels. Follow-up experiments revealed that PrP collaborates with members of the MARCKS protein family in its control of NCAM1 polysialylation. We conclude that the physiological function of PrP may be masked in analyses of complex brain samples but its cell-type specific influence on a lipid raft-based NCAM1-related cell biology comes to the fore in investigations of specific cell types.


Assuntos
Encéfalo/metabolismo , Modelos Biológicos , Proteínas Priônicas/deficiência , Proteoma/metabolismo , Sequência de Aminoácidos , Animais , Antígeno CD56/metabolismo , Proteínas de Ligação a Calmodulina , Linhagem Celular , Análise por Conglomerados , Ontologia Genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos , Substrato Quinase C Rico em Alanina Miristoilada , Ácido N-Acetilneuramínico/metabolismo , Proteínas Priônicas/metabolismo , Proteômica , Reprodutibilidade dos Testes , Homologia de Sequência de Aminoácidos , Transdução de Sinais
11.
Mol Cell Proteomics ; 14(11): 3000-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26269332

RESUMO

The tau protein is central to the etiology of several neurodegenerative diseases, including Alzheimer's disease, a subset of frontotemporal dementias, progressive supranuclear palsy and dementia following traumatic brain injury, yet the proteins it interacts with have not been studied using a systematic discovery approach. Here we employed mild in vivo crosslinking, isobaric labeling, and tandem mass spectrometry to characterize molecular interactions of human tau in a neuroblastoma cell model. The study revealed a robust association of tau with the ribonucleoproteome, including major protein complexes involved in RNA processing and translation, and documented binding of tau to several heat shock proteins, the proteasome and microtubule-associated proteins. Follow-up experiments determined the relative contribution of cellular RNA to the tau interactome and mapped interactions to N- or C-terminal tau domains. We further document that expression of P301L mutant tau disrupts interactions of the C-terminal half of tau with heat shock proteins and the proteasome. The data are consistent with a model whereby a higher propensity of P301L mutant tau to aggregate may reflect a perturbation of its chaperone-assisted stabilization and proteasome-dependent degradation. Finally, using a global proteomics approach, we show that heterologous expression of a tau construct that lacks the C-terminal domain, including the microtubule binding domain, does not cause a discernible shift of the proteome except for a significant direct correlation of steady-state levels of tau and cystatin B.


Assuntos
Células Epiteliais/metabolismo , Chaperonas Moleculares/metabolismo , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas tau/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Cistatina B/genética , Cistatina B/metabolismo , Células Epiteliais/citologia , Regulação da Expressão Gênica , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Anotação de Sequência Molecular , Mutação , Neurônios/citologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Ribonucleoproteínas/genética , Transdução de Sinais , Proteínas tau/genética
12.
PLoS One ; 10(8): e0133741, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26288071

RESUMO

Despite its multi-faceted role in neurodegenerative diseases, the physiological function of the prion protein (PrP) has remained elusive. On the basis of its evolutionary relationship to ZIP metal ion transporters, we considered that PrP may contribute to the morphogenetic reprogramming of cells underlying epithelial-to-mesenchymal transitions (EMT). Consistent with this hypothesis, PrP transcription increased more than tenfold during EMT, and stable PrP-deficient cells failed to complete EMT in a mammalian cell model. A global comparative proteomics analysis identified the neural cell adhesion molecule 1 (NCAM1) as a candidate mediator of this impairment, which led to the observation that PrP-deficient cells fail to undergo NCAM1 polysialylation during EMT. Surprisingly, this defect was caused by a perturbed transcription of the polysialyltransferase ST8SIA2 gene. Proteomics data pointed toward ß-catenin as a transcriptional regulator affected in PrP-deficient cells. Indeed, pharmacological blockade or siRNA-based knockdown of ß-catenin mimicked PrP-deficiency in regards to NCAM1 polysialylation. Our data established the existence of a PrP-ST8SIA2-NCAM signaling loop, merged two mature fields of investigation and offer a simple model for explaining phenotypes linked to PrP.


Assuntos
Antígeno CD56/metabolismo , Morfogênese/fisiologia , Príons/metabolismo , Animais , Linhagem Celular , Transição Epitelial-Mesenquimal/fisiologia , Camundongos , Proteômica/métodos , Sialiltransferases/metabolismo , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia , beta Catenina/metabolismo
13.
PLoS One ; 9(12): e114594, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25490046

RESUMO

The molecular function of the cellular prion protein (PrPC) and the mechanism by which it may contribute to neurotoxicity in prion diseases and Alzheimer's disease are only partially understood. Mouse neuroblastoma Neuro2a cells and, more recently, C2C12 myocytes and myotubes have emerged as popular models for investigating the cellular biology of PrP. Mouse epithelial NMuMG cells might become attractive models for studying the possible involvement of PrP in a morphogenetic program underlying epithelial-to-mesenchymal transitions. Here we describe the generation of PrP knockout clones from these cell lines using CRISPR-Cas9 knockout technology. More specifically, knockout clones were generated with two separate guide RNAs targeting recognition sites on opposite strands within the first hundred nucleotides of the Prnp coding sequence. Several PrP knockout clones were isolated and genomic insertions and deletions near the CRISPR-target sites were characterized. Subsequently, deep quantitative global proteome analyses that recorded the relative abundance of>3000 proteins (data deposited to ProteomeXchange Consortium) were undertaken to begin to characterize the molecular consequences of PrP deficiency. The levels of ∼ 120 proteins were shown to reproducibly correlate with the presence or absence of PrP, with most of these proteins belonging to extracellular components, cell junctions or the cytoskeleton.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Príons/genética , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Camundongos , Proteoma
14.
Front Cell Dev Biol ; 2: 53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25453033

RESUMO

Knowledge of phenotypic changes the cellular prion protein (PrP(C)) contributes to may provide novel avenues for understanding its function. Here we consider data from functional knockout/down studies and protein-protein interaction analyses from the perspective of PrP's relationship to its ancestral ZIP metal ion transporting proteins. When approached in this manner, a role of PrP(C) as a modulator of a complex morphogenetic program that underlies epithelial-to-mesenchymal transition (EMT) emerges. To execute EMT, cells have to master the challenge to shift from cell-cell to cell-substrate modes of adherence. During this process, cell-cell junctions stabilized by E-cadherins are replaced by focal adhesions that mediate cell-substrate contacts. A similar reprogramming occurs during distinct organogenesis events that have been shown to rely on ZIP transporters. A model is presented that sees ZIP transporters, and possibly also PrP(C), affect this balance of adherence modes at both the transcriptional and post-translational levels.

15.
PLoS One ; 8(9): e72446, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039764

RESUMO

The cellular prion protein (PrP(C)) was recently observed to co-purify with members of the LIV-1 subfamily of ZIP zinc transporters (LZTs), precipitating the surprising discovery that the prion gene family descended from an ancestral LZT gene. Here, we compared the subcellular distribution and biophysical characteristics of LZTs and their PrP-like ectodomains. When expressed in neuroblastoma cells, the ZIP5 member of the LZT subfamily was observed to be largely directed to the same subcellular locations as PrP(C) and both proteins were seen to be endocytosed through vesicles decorated with the Rab5 marker protein. When recombinantly expressed, the PrP-like domain of ZIP5 could be obtained with yields and levels of purity sufficient for structural analyses but it tended to aggregate, thereby precluding attempts to study its structure. These obstacles were overcome by moving to a mammalian cell expression system. The subsequent biophysical characterization of a homogeneous preparation of the ZIP5 PrP-like ectodomain shows that this protein acquires a dimeric, largely globular fold with an α-helical content similar to that of mammalian PrP(C). The use of a mammalian cell expression system also allowed for the expression and purification of stable preparations of Takifugu rubripes PrP-1, thereby overcoming a key hindrance to high-resolution work on a fish PrP(C).


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas PrPC/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte de Cátions/química , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Endossomos/metabolismo , Células HEK293 , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas rab5 de Ligação ao GTP/metabolismo
16.
J Neurol ; 260(3): 856-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23108490

RESUMO

Spinocerebellar ataxias (SCA) are a genetically heterogeneous group of neurodegenerative diseases characterised by progressive cerebellar ataxia, dysarthria and oculomotor abnormalities. Recently the prodynorphin (PDYN) gene was identified as the cause of SCA23 in four Dutch families displaying progressive gait and limb ataxia. In this study we aimed to assess the frequency of PDYN gene defects and extend the phenotype of SCA23 patients in a UK ataxia series and also in patients from Greece, Egypt and India. We sequenced the coding and flanking intronic regions of the PDYN gene in a total of 852 ataxia patients, of which 356 were sporadic with no family history, 320 had a positive family history, and 176 probands had a positive family history and at least one family member had also been investigated. We also analysed 190 patients with multiple-system atrophy with cerebellar features (MSA-C), a phenocopy of SCA23. We identified a novel putative pathogenic heterozygous missense variant in the PDYN gene in an early onset SCA patient with an unknown family history. This variant was not present in 570 matched British controls. This is the first study to screen for SCA23 in UK patients and confirms that PDYN mutations are a very rare cause of spinocerebellar ataxia, accounting for ~ 0.1 % of ataxia cases but perhaps with a higher frequency in pure cerebellar ataxia. Given the rarity of PDYN mutations, front-line diagnostic evaluation of UK familial and early onset pure spinocerebellar ataxia patients should focus on other known ataxia genes.


Assuntos
Encefalinas/genética , Vigilância da População , Precursores de Proteínas/genética , Degenerações Espinocerebelares/epidemiologia , Degenerações Espinocerebelares/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vigilância da População/métodos , Prevalência , Degenerações Espinocerebelares/diagnóstico , Reino Unido/epidemiologia
17.
J Mol Biol ; 422(4): 556-574, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22687393

RESUMO

We recently documented the co-purification of members of the LIV-1 subfamily of ZIP (Zrt-, Irt-like Protein) zinc transporters (LZTs) with the cellular prion protein (PrP(C)) and, subsequently, established that the prion gene family descended from an ancestral LZT gene. Here, we begin to address whether the study of LZTs can shed light on the biology of prion proteins in health and disease. Starting from an observation of an abnormal LZT immunoreactive band in prion-infected mice, subsequent cell biological analyses uncovered a surprisingly coordinated biology of ZIP10 (an LZT member) and prion proteins that involves alterations to N-glycosylation and endoproteolysis in response to manipulations to the extracellular divalent cation milieu. Starving cells of manganese or zinc, but not copper, causes shedding of the N1 fragment of PrP(C) and of the ectodomain of ZIP10. For ZIP10, this posttranslational biology is influenced by an interaction between its PrP-like ectodomain and a conserved metal coordination site within its C-terminal multi-spanning transmembrane domain. The transition metal starvation-induced cleavage of ZIP10 can be differentiated by an immature N-glycosylation signature from a constitutive cleavage targeting the same site. Data from this work provide a first glimpse into a hitherto neglected molecular biology that ties PrP to its LZT cousins and suggest that manganese or zinc starvation may contribute to the etiology of prion disease in mice.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Metais/metabolismo , Príons/metabolismo , Elementos de Transição/metabolismo , Sequência de Aminoácidos , Animais , Cátions Bivalentes/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Glicosilação , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Doenças Priônicas/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteólise , Zinco/metabolismo
18.
Prion ; 6(4): 317-21, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22575750

RESUMO

The evolutionary origins of vertebrate prion genes had remained elusive until recently when multiple lines of evidence converged to the proposition that members of the prion gene family represent an ancient branch of a larger family of ZIP metal ion transporters. (1) A follow-up investigation which explored the mechanism of evolution in more detail led to the surprising conclusion that the emergence of the prion founder gene likely involved the reverse transcription of a spliced transcript of a LIV-1 ZIP predecessor gene. (2) The objective of this perspective is to discuss the possible significance of this reunion of ZIP and prion gene subfamilies for understanding the biology of the prion protein in health and disease. While a recent review article broadly introduced this area of research, (3) the emphasis here is to comment on some of the more pertinent concepts, experimental paradigms, ongoing developments and challenges.


Assuntos
Proteínas de Transporte de Cátions/genética , Príons/genética , Animais , Proteínas de Transporte de Cátions/análise , Proteínas de Transporte de Cátions/metabolismo , Evolução Molecular , Humanos , Modelos Moleculares , Príons/análise , Príons/metabolismo , Zinco/metabolismo
19.
Chem Biol Drug Des ; 79(2): 166-76, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21974743

RESUMO

Defining the role of calcitonin gene-related peptide in migraine pathogenesis could lead to the application of calcitonin gene-related peptide antagonists as novel migraine therapeutics. In this work, quantitative structure-activity relationship modeling of biological activities of a large range of calcitonin gene-related peptide antagonists was performed using a panel of physicochemical descriptors. The computational studies evaluated different variable selection techniques and demonstrated shuffling stepwise multiple linear regression to be superior over genetic algorithm-multiple linear regression. The linear quantitative structure-activity relationship model revealed better statistical parameters of cross-validation in comparison with the non-linear support vector regression technique. Implementing only five peptide descriptors into this linear quantitative structure-activity relationship model resulted in an extremely robust and highly predictive model with calibration, leave-one-out and leave-20-out validation R(2) of 0.9194, 0.9103, and 0.9214, respectively. We performed docking of the most potent calcitonin gene-related peptide antagonists with the calcitonin gene-related peptide receptor and demonstrated that peptide antagonists act by blocking access to the peptide-binding cleft. We also demonstrated the direct contact of residues 28-37 of the calcitonin gene-related peptide antagonists with the receptor. These results are in agreement with the conclusions drawn from the quantitative structure-activity relationship model, indicating that both electrostatic and steric factors should be taken into account when designing novel calcitonin gene-related peptide antagonists.


Assuntos
Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina , Modelos Moleculares , Relação Quantitativa Estrutura-Atividade , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Análise de Componente Principal , Estrutura Terciária de Proteína , Proteína 1 Modificadora da Atividade de Receptores/antagonistas & inibidores , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/química , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo , Software , Eletricidade Estática
20.
Artigo em Inglês | MEDLINE | ID: mdl-23439843

RESUMO

BACKGROUND: Hereditary spastic paraplegias (HSPs) are a clinically and genetically heterogeneous group of neurological diseases, which typically present with progressive lower extremity weakness and spasticity causing progressive walking difficulties. Complicating neurological or extraneurological features may be present. CASE REPORT: We describe a 19-year-old male who was referred because of an action tremor of the hands; he later developed walking difficulties. Callosal atrophy was present on his cerebral magnetic resonance imaging scan, prompting genetic testing for SPG11, which revealed homozygous mutations. DISCUSSION: The clinical features, differential diagnosis and management of SPG11, the most common form of autosomal recessive complicated HSP with a thin corpus callosum are discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...