Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Meat Sci ; 195: 109008, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36274373

RESUMEN

This study evaluated whether different colour intensities of ribeye steaks from dark-cutting (B4 grade) beef carcasses (Dark-B4DK/Moderate-B4MD) were similar in appearance and eating quality to steaks from normal (N) carcasses of lower marbling grades (AA or A) as assessed by consumers. The B4MDAAA and B4DK/MDAA had similar raw appearance and eating quality to N carcasses with a one quality downgrade for marbling (P > 0.1), potentially supporting a B4MDAAA and B4DK/MDAA re-class to NAA and NA grades, respectively. Cooked B4DKAAA steaks had greater juiciness and tenderness acceptability (P < 0.01) and similar appearance, flavour and overall acceptability and purchase intent compared to NAA steaks (P > 0.1). However, consumers perceived greater marbling and lower colour acceptability (P < 0.01) in raw B4DKAAA compared to NAA steaks, lowering the purchase intent scores of B4DKAAA steaks (P < 0.01). These results suggest merit for continuing a B4DKAAA segregated grade, unless the superior eating quality of B4DKAAA could offset its poorer raw appearance through consumer education or modified atmosphere packaging.


Asunto(s)
Comportamiento del Consumidor , Carne , Animales , Bovinos , Canadá , Culinaria , Gusto
2.
Exp Neurol ; 328: 113283, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32165257

RESUMEN

It has been proposed that Amyloid ß Precursor Protein (APP) might act as a rheostat controlling neuronal excitability, but mechanisms have remained untested. APP and its catabolite Aß are known to impact upon synapse function and dysfunction via their interaction with the prion protein (PrPC), suggesting a candidate pathway. Here we test if PrPC is required for this APP function in vivo, perhaps via modulating mGluR5 ion channels. We engineered zebrafish to lack homologs of PrPC and APP, allowing us to assess their purported genetic and physiological interactions in CNS development. We generated four appa null alleles as well as prp1-/-;appa-/- double mutants (engineering of prp1 mutant alleles is described elsewhere). Unexpectedly, appa-/- and compound prp1-/-;appa-/- mutants are viable and lacked overt phenotypes (except being slightly smaller than wildtype fish at some developmental stages). Zebrafish prp1-/- mutants were substantially more sensitive to appa knockdown than wildtype fish, and both zebrafish prp1 and mammalian Prnp mRNA were significantly able to partially rescue this effect. Further, appa-/- mutants exhibited increased seizures upon exposure to low doses of convulsant. The mechanism of this seizure susceptibility requires prp1 insomuch that seizures were significantly dampened to wildtype levels in prp1-/-;appa-/- mutants. Inhibiting mGluR5 channels, which may be downstream of PrPC, increased seizure intensity only in prp1-/- mutants, and this seizure mechanism required intact appa. Taken together, these results support an intriguing genetic interaction between prp1 and appa with their shared roles impacting upon neuron hyperexcitability, thus complementing and extending past works detailing their biochemical interaction(s).


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Susceptibilidad a Enfermedades/metabolismo , Proteínas Priónicas/metabolismo , Convulsiones/genética , Convulsiones/metabolismo , Animales , Ratones , Mutación , Pez Cebra
3.
J Biol Chem ; 293(32): 12576-12592, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-29903907

RESUMEN

Normally folded prion protein (PrPC) and its functions in healthy brains remain underappreciated compared with the intense study of its misfolded forms ("prions," PrPSc) during the pathobiology of prion diseases. This impedes the development of therapeutic strategies in Alzheimer's and prion diseases. Disrupting the zebrafish homologs of PrPC has provided novel insights; however, mutagenesis of the zebrafish paralog prp2 did not recapitulate previous dramatic developmental phenotypes, suggesting redundancy with the prp1 paralog. Here, we generated zebrafish prp1 loss-of-function mutant alleles and dual prp1-/-;prp2-/- mutants. Zebrafish prp1-/- and dual prp1-/-;prp2-/- mutants resemble mammalian Prnp knockouts insofar as they lack overt phenotypes, which surprisingly contrasts with reports of severe developmental phenotypes when either prp1 or prp2 is knocked down acutely. Previous studies suggest that PrPC participates in neural cell development/adhesion, including in zebrafish where loss of prp2 affects adhesion and deposition patterns of lateral line neuromasts. In contrast with the expectation that prp1's functions would be redundant to prp2, they appear to have opposing functions in lateral line neurodevelopment. Similarly, loss of prp1 blunted the seizure susceptibility phenotypes observed in prp2 mutants, contrasting the expected exacerbation of phenotypes if these prion gene paralogs were serving redundant roles. In summary, prion mutant fish lack the overt phenotypes previously predicted, and instead they have subtle phenotypes similar to mammals. No evidence was found for functional redundancy in the zebrafish prion gene paralogs, and the phenotypes observed when each gene is disrupted individually are consistent with ancient functions of prion proteins in neurodevelopment and modulation of neural activity.


Asunto(s)
Animales Modificados Genéticamente/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Enfermedades por Prión/fisiopatología , Proteínas Priónicas/genética , Convulsiones/fisiopatología , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente/genética , Mutación , Fenotipo , Pez Cebra/genética
4.
Biol Open ; 7(1)2018 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-29358166

RESUMEN

The misfolding of cellular prion protein (PrPC) to form PrP Scrapie (PrPSc) is an exemplar of toxic gain-of-function mechanisms inducing propagated protein misfolding and progressive devastating neurodegeneration. Despite this, PrPC function in the brain is also reduced and subverted during prion disease progression; thus understanding the normal function of PrPC in healthy brains is key. Disrupting PrPC in mice has led to a myriad of controversial functions that sometimes map onto disease symptoms, including a proposed role in memory or learning. Intriguingly, PrPC interaction with amyloid beta (Aß) oligomers at synapses has also linked its function to Alzheimer's disease and dementia in recent years. We set out to test the involvement of PrPC in memory using a disparate animal model, the zebrafish. Here we document an age-dependent memory decline in prp2-/- zebrafish, pointing to a conserved and ancient role of PrPC in memory. Specifically, we found that aged (3-year-old) prp2-/- fish performed poorly in an object recognition task relative to age-matched prp2+/+ fish or 1-year-old prp2-/- fish. Further, using a novel object approach (NOA) test, we found that aged (3-year-old) prp2-/- fish approached the novel object more than either age-matched prp2+/+ fish or 1-year-old prp2-/- fish, but did not have decreased anxiety when we tested them in a novel tank diving test. Taken together, the results of the NOA and novel tank diving tests suggest an altered cognitive appraisal of the novel object in the 3-year-old prp2-/- fish. The learning paradigm established here enables a path forward to study PrPC interactions of relevance to Alzheimer's disease and prion diseases, and to screen for candidate therapeutics for these diseases. The findings underpin a need to consider the relative contributions of loss- versus gain-of-function of PrPC during Alzheimer's and prion diseases, and have implications upon the prospects of several promising therapeutic strategies.

5.
Int J Mol Sci ; 18(10)2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29064456

RESUMEN

Prions have served as pathfinders that reveal many aspects of proteostasis in neurons. The recent realization that several prominent neurodegenerative diseases spread via a prion-like mechanism illuminates new possibilities for diagnostics and therapeutics. Thus, key proteins in Alzheimer Disease and Amyotrophic lateral sclerosis (ALS), including amyloid-ß precursor protein, Tau and superoxide dismutase 1 (SOD1), spread to adjacent cells in their misfolded aggregated forms and exhibit template-directed misfolding to induce further misfolding, disruptions to proteostasis and toxicity. Here we invert this comparison to ask what these prion-like diseases can teach us about the broad prion disease class, especially regarding the loss of these key proteins' function(s) as they misfold and aggregate. We also consider whether functional amyloids might reveal a role for subverted protein function in neurodegenerative disease. Our synthesis identifies SOD1 as an exemplar of protein functions being lost during prion-like protein misfolding, because SOD1 is inherently unstable and loses function in its misfolded disease-associated form. This has under-appreciated parallels amongst the canonical prion diseases, wherein the normally folded prion protein, PrPC, is reduced in abundance in fatal familial insomnia patients and during the preclinical phase in animal models, apparently via proteostatic mechanisms. Thus while template-directed misfolding and infectious properties represent gain-of-function that fascinates proteostasis researchers and defines (is required for) the prion(-like) diseases, loss and subversion of the functions attributed to hallmark proteins in neurodegenerative disease needs to be integrated into design towards effective therapeutics. We propose experiments to uniquely test these ideas.


Asunto(s)
Enfermedades por Prión/patología , Proteínas Priónicas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Humanos , Enfermedades por Prión/metabolismo , Proteínas Priónicas/química , Pliegue de Proteína , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo
6.
J Alzheimers Dis ; 54(1): 3-29, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27392869

RESUMEN

Prion disease research has contributed much toward understanding other neurodegenerative diseases, including recent demonstrations that Alzheimer's disease (AD) and other neurodegenerative diseases are prion-like. Prion-like diseases involve the spread of degeneration between individuals and/or among cells or tissues via template directed misfolding, wherein misfolded protein conformers propagate disease by causing normal proteins to misfold. Here we use the premise that AD, amyotrophic lateral sclerosis, Huntington's disease, and other similar diseases are prion-like and ask: Can we apply knowledge gained from studies of these prion-like diseases to resolve debates about classical prion diseases? We focus on controversies about what role(s) protein loss-of-function might have in prion diseases because this has therapeutic implications, including for AD. We examine which loss-of-function events are recognizable in prion-like diseases by considering the normal functions of the proteins before their misfolding and aggregation. We then delineate scenarios wherein gain-of-function and/or loss-of-function would be necessary or sufficient for neurodegeneration. We consider roles of PrPC loss-of-function in prion diseases and in AD, and conclude that the conventional wisdom that prion diseases are 'toxic gain-of-function diseases' has limitations. While prion diseases certainly have required gain-of-function components, we propose that disease phenotypes are predominantly caused by deficits in the normal physiology of PrPC and its interaction partners as PrPC converts to PrPSc. In this model, gain-of-function serves mainly to spread disease, and loss-of-function directly mediates neuron dysfunction. We propose experiments and predictions to assess our conclusion. Further study on the normal physiological roles of these key proteins is warranted.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Animales , Humanos , Pliegue de Proteína
7.
PLoS One ; 9(12): e113331, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25436888

RESUMEN

Prion protein is involved in severe neurodegenerative disorders but its physiological role is still in debate due to an absence of major developmental defects in knockout mice. Previous reports in zebrafish indicate that the two prion genes, PrP1 and PrP2, are both involved in several steps of embryonic development thus providing a unique route to discover prion protein function. Here we investigate the role of PrP2 during development of a mechano-sensory system, the posterior lateral line, using morpholino knockdown and PrP2 targeted inactivation. We confirm the efficiency of the translation blocking morpholino at the protein level. Development of the posterior lateral line is altered in PrP2 morphants, including nerve axonal outgrowth and primordium migration defects. Reduced neuromast deposition was observed in PrP2 morphants as well as in PrP2-/- mutants. Rosette formation defects were observed in PrP2 morphants, strongly suggesting an abnormal primordium organization and reflecting loss of cell cohesion during migration of the primordium. In addition, the adherens junction proteins, E-cadherin and ß-catenin, were mis-localized after reduction of PrP2 expression and thus contribute to the primordium disorganization. Consequently, hair cell differentiation and number were affected and this resulted in reduced functional neuromasts. At later developmental stages, myelination of the posterior lateral line nerve was altered. Altogether, our study reports an essential role of PrP2 in collective migration process of the primordium and in neuromast formation, further implicating a role for prion protein in cell adhesion.


Asunto(s)
Movimiento Celular , Mecanorreceptores/citología , Priones/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Uniones Adherentes/metabolismo , Animales , Axones/metabolismo , Adhesión Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Células Ciliadas Auditivas/citología , Humanos , Mecanorreceptores/metabolismo , Ratones , Priones/genética , Células de Schwann/citología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Zebrafish ; 10(3): 353-64, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23781947

RESUMEN

Synthetic targeted endonucleases such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) have recently emerged as powerful tools for targeted mutagenesis, especially in organisms that are not amenable to embryonic stem cell manipulation. Both ZFNs and TALENs consist of DNA-binding arrays that are fused to the nonspecific FokI nuclease domain. In an effort to improve targeted endonuclease mutagenesis efficiency, we enhanced their catalytic activity using the Sharkey FokI nuclease domain variant. All constructs tested display increased DNA cleavage activity in vitro. We demonstrate that one out of four ZFN arrays containing the Sharkey FokI variant exhibits a dramatic increase in mutagenesis frequency in vivo in zebrafish. The other three ZFNs exhibit no significant alteration of activity in vivo. Conversely, we demonstrate that TALENs containing the Sharkey FokI variant exhibit absent or severely reduced in vivo mutagenic activity in zebrafish. Notably, Sharkey ZFNs and TALENs do not generate increased toxicity-related defects or mortality. Our results present Sharkey ZFNs as an effective alternative to conventional ZFNs, but advise against the use of Sharkey TALENs.


Asunto(s)
Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Pez Cebra/genética , Animales , Dedos de Zinc
9.
Neurobiol Dis ; 55: 11-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23523635

RESUMEN

The function of the cellular prion protein (PrP(C)) in healthy brains remains poorly understood, in part because Prnp knockout mice are viable. On the other hand, transient knockdown of Prnp homologs in zebrafish (including two paralogs, prp1 and prp2) has suggested that PrP(C) is required for CNS development, cell adhesion, and neuroprotection. It has been argued that zebrafish Prp2 is most similar to mammalian PrP(C), yet it has remained intransigent to the most thorough confirmations of reagent specificity during knockdown. Thus we investigated the role of prp2 using targeted gene disruption via zinc finger nucleases. Prp2(-/-) zebrafish were viable and did not display overt developmental phenotypes. Back-crossing female prp2(-/-) fish ruled out a role for maternal mRNA contributions. Prp2(-/-) larvae were found to have increased seizure-like behavior following exposure to the convulsant pentylenetetrazol (PTZ), as compared to wild type fish. In situ recordings from intact hindbrains demonstrated that prp2 regulates closing of N-Methyl-d-aspartate (NMDA) receptors, concomitant with neuroprotection during glutamate excitotoxicity. Overall, the knockout of Prp2 function in zebrafish independently confirmed hypothesized roles for PrP, identifying deeply conserved functions in post-developmental regulation of neuron excitability that are consequential to the etiology of prion and Alzheimer diseases.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Mutación/genética , Neuronas/metabolismo , Priones/genética , Factores de Edad , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/fisiopatología , Biblioteca de Genes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Larva , Ratones , Mutagénesis Sitio-Dirigida , Pentilenotetrazol/toxicidad , Fenotipo , Receptores de N-Metil-D-Aspartato/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Dedos de Zinc/genética
10.
PLoS One ; 7(12): e51305, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23236467

RESUMEN

Genetic and biochemical mechanisms linking onset or progression of Alzheimer Disease and prion diseases have been lacking and/or controversial, and their etiologies are often considered independent. Here we document a novel, conserved and specific genetic interaction between the proteins that underlie these diseases, amyloid-ß precursor protein and prion protein, APP and PRP, respectively. Knockdown of APP and/or PRNP homologs in the zebrafish (appa, appb, prp1, and prp2) produces a dose-dependent phenotype characterized by systemic morphological defects, reduced cell adhesion and CNS cell death. This genetic interaction is surprisingly exclusive in that prp1 genetically interacts with zebrafish appa, but not with appb, and the zebrafish paralog prp2 fails to interact with appa. Intriguingly, appa & appb are largely redundant in early zebrafish development yet their abilities to rescue CNS cell death are differentially contingent on prp1 abundance. Delivery of human APP or mouse Prnp mRNAs rescue the phenotypes observed in app-prp-depleted zebrafish, highlighting the conserved nature of this interaction. Immunoprecipitation revealed that human APP and PrP(C) proteins can have a physical interaction. Our study reports a unique in vivo interdependence between APP and PRP loss-of-function, detailing a biochemical interaction that considerably expands the hypothesized roles of PRP in Alzheimer Disease.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Apoptosis/fisiología , Adhesión Celular/fisiología , Sistema Nervioso Central/embriología , Fenotipo , Priones/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/genética , Sistema Nervioso Central/citología , Clonación Molecular , Cartilla de ADN/genética , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Ratones , Morfolinos/genética , Mutagénesis Sitio-Dirigida , Priones/genética , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA