Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gene Ther ; 31(5-6): 234-241, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38135787

RESUMEN

EPM1 is the most common form of Progressive Myoclonus Epilepsy characterized by late-childhood onset, ever-worsening and disabling myoclonus, seizures, ataxia, psychiatric disease, and shortened lifespan. EPM1 is caused by expansions of a dodecamer repeat sequence in the promoter of CSTB (cystatin B), which dramatically reduces, but does not eliminate, gene expression. The relatively late onset and consistent presence of a minimal amount of protein product makes EPM1 a favorable target for gene replacement therapy. If treated early, these children's normally developed brains could be rescued from the neurodegeneration that otherwise follows, and their cross-reactive immunological material (CRIM) positive status greatly reduces transgene related toxicity. We performed a proof-of-concept CSTB gene replacement study in Cstb knockout mice by introducing full-length human CSTB driven by the CBh promoter packaged in AAV9 and administered at postnatal days 21 and 60. Mice were sacrificed at 2 or 9 months of age, respectively. We observed significant improvements in expression levels of neuroinflammatory pathway genes and cerebellar granule cell layer apoptosis, as well as amelioration of motor impairment. The data suggest that gene replacement is a promising therapeutic modality for EPM1 and could spare affected children and families the ravages of this otherwise severe neurodegenerative disease.


Asunto(s)
Cistatina B , Terapia Genética , Ratones Noqueados , Enfermedades Neuroinflamatorias , Animales , Ratones , Terapia Genética/métodos , Cistatina B/genética , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/genética , Humanos , Ataxia/genética , Ataxia/terapia , Epilepsias Mioclónicas Progresivas/genética , Epilepsias Mioclónicas Progresivas/terapia , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación
2.
Acta Neuropathol ; 147(1): 46, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-38411740

RESUMEN

At least five enzymes including three E3 ubiquitin ligases are dedicated to glycogen's spherical structure. Absence of any reverts glycogen to a structure resembling amylopectin of the plant kingdom. This amylopectinosis (polyglucosan body formation) causes fatal neurological diseases including adult polyglucosan body disease (APBD) due to glycogen branching enzyme deficiency, Lafora disease (LD) due to deficiencies of the laforin glycogen phosphatase or the malin E3 ubiquitin ligase and type 1 polyglucosan body myopathy (PGBM1) due to RBCK1 E3 ubiquitin ligase deficiency. Little is known about these enzymes' functions in glycogen structuring. Toward understanding these functions, we undertake a comparative murine study of the amylopectinoses of APBD, LD and PGBM1. We discover that in skeletal muscle, polyglucosan bodies form as two main types, small and multitudinous ('pebbles') or giant and single ('boulders'), and that this is primarily determined by the myofiber types in which they form, 'pebbles' in glycolytic and 'boulders' in oxidative fibers. This pattern recapitulates what is known in the brain in LD, innumerable dust-like in astrocytes and single giant sized in neurons. We also show that oxidative myofibers are relatively protected against amylopectinosis, in part through highly increased glycogen branching enzyme expression. We present evidence of polyglucosan body size-dependent cell necrosis. We show that sex influences amylopectinosis in genotype, brain region and myofiber-type-specific fashion. RBCK1 is a component of the linear ubiquitin chain assembly complex (LUBAC), the only known cellular machinery for head-to-tail linear ubiquitination critical to numerous cellular pathways. We show that the amylopectinosis of RBCK1 deficiency is not due to loss of linear ubiquitination, and that another function of RBCK1 or LUBAC must exist and operate in the shaping of glycogen. This work opens multiple new avenues toward understanding the structural determinants of the mammalian carbohydrate reservoir critical to neurologic and neuromuscular function and disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo IV , Enfermedad del Almacenamiento de Glucógeno , Enfermedades del Sistema Nervioso , Animales , Ratones , Glucógeno , Ubiquitina-Proteína Ligasas , Ubiquitinas , Mamíferos
3.
Epilepsia ; 65(3): 709-724, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38231304

RESUMEN

OBJECTIVE: KCTD7-related progressive myoclonic epilepsy (PME) is a rare autosomal-recessive disorder. This study aimed to describe the clinical details and genetic variants in a large international cohort. METHODS: Families with molecularly confirmed diagnoses of KCTD7-related PME were identified through international collaboration. Furthermore, a systematic review was done to identify previously reported cases. Salient demographic, epilepsy, treatment, genetic testing, electroencephalographic (EEG), and imaging-related variables were collected and summarized. RESULTS: Forty-two patients (36 families) were included. The median age at first seizure was 14 months (interquartile range = 11.75-22.5). Myoclonic seizures were frequently the first seizure type noted (n = 18, 43.9%). EEG and brain magnetic resonance imaging findings were variable. Many patients exhibited delayed development with subsequent progressive regression (n = 16, 38.1%). Twenty-one cases with genetic testing available (55%) had previously reported variants in KCTD7, and 17 cases (45%) had novel variants in KCTD7 gene. Six patients died in the cohort (age range = 1.5-21 years). The systematic review identified 23 eligible studies and further identified 59 previously reported cases of KCTD7-related disorders from the literature. The phenotype for the majority of the reported cases was consistent with a PME (n = 52, 88%). Other reported phenotypes in the literature included opsoclonus myoclonus ataxia syndrome (n = 2), myoclonus dystonia (n = 2), and neuronal ceroid lipofuscinosis (n = 3). Eight published cases died over time (14%, age range = 3-18 years). SIGNIFICANCE: This study cohort and systematic review consolidated the phenotypic spectrum and natural history of KCTD7-related disorders. Early onset drug-resistant epilepsy, relentless neuroregression, and severe neurological sequalae were common. Better understanding of the natural history may help future clinical trials.


Asunto(s)
Epilepsias Mioclónicas , Epilepsias Mioclónicas Progresivas , Síndrome de Unverricht-Lundborg , Adolescente , Niño , Preescolar , Humanos , Lactante , Adulto Joven , Electroencefalografía , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas Progresivas/genética , Canales de Potasio/genética , Convulsiones
4.
Cell ; 137(2): 235-46, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19379691

RESUMEN

X-linked myopathy with excessive autophagy (XMEA) is a childhood-onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p it is an essential assembly chaperone of the V-ATPase, the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH, which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids, which upregulates the mTOR pathway and mTOR-dependent macroautophagy, resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge together, and vacuolate the cell. Our results uncover macroautophagic overcompensation leading to cell vacuolation and tissue atrophy as a mechanism of disease.


Asunto(s)
Genes Ligados a X , Enfermedades Musculares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Autofagia , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , ARN Mensajero/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética
5.
Brain ; 145(7): 2361-2377, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35084461

RESUMEN

Longer glucan chains tend to precipitate. Glycogen, by far the largest mammalian glucan and the largest molecule in the cytosol with up to 55 000 glucoses, does not, due to a highly regularly branched spherical structure that allows it to be perfused with cytosol. Aberrant construction of glycogen leads it to precipitate, accumulate into polyglucosan bodies that resemble plant starch amylopectin and cause disease. This pathology, amylopectinosis, is caused by mutations in a series of single genes whose functions are under active study toward understanding the mechanisms of proper glycogen construction. Concurrently, we are characterizing the physicochemical particularities of glycogen and polyglucosans associated with each gene. These genes include GBE1, EPM2A and EPM2B, which respectively encode the glycogen branching enzyme, the glycogen phosphatase laforin and the laforin-interacting E3 ubiquitin ligase malin, for which an unequivocal function is not yet known. Mutations in GBE1 cause a motor neuron disease (adult polyglucosan body disease), and mutations in EPM2A or EPM2B a fatal progressive myoclonus epilepsy (Lafora disease). RBCK1 deficiency causes an amylopectinosis with fatal skeletal and cardiac myopathy (polyglucosan body myopathy 1, OMIM# 615895). RBCK1 is a component of the linear ubiquitin chain assembly complex, with unique functions including generating linear ubiquitin chains and ubiquitinating hydroxyl (versus canonical amine) residues, including of glycogen. In a mouse model we now show (i) that the amylopectinosis of RBCK1 deficiency, like in adult polyglucosan body disease and Lafora disease, affects the brain; (ii) that RBCK1 deficiency glycogen, like in adult polyglucosan body disease and Lafora disease, has overlong branches; (iii) that unlike adult polyglucosan body disease but like Lafora disease, RBCK1 deficiency glycogen is hyperphosphorylated; and finally (iv) that unlike laforin-deficient Lafora disease but like malin-deficient Lafora disease, RBCK1 deficiency's glycogen hyperphosphorylation is limited to precipitated polyglucosans. In summary, the fundamental glycogen pathology of RBCK1 deficiency recapitulates that of malin-deficient Lafora disease. Additionally, we uncover sex and genetic background effects in RBCK1 deficiency on organ- and brain-region specific amylopectinoses, and in the brain on consequent neuroinflammation and behavioural deficits. Finally, we exploit the portion of the basic glycogen pathology that is common to adult polyglucosan body disease, both forms of Lafora disease and RBCK1 deficiency, namely overlong branches, to show that a unified approach based on downregulating glycogen synthase, the enzyme that elongates glycogen branches, can rescue all four diseases.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo IV , Enfermedad de Lafora , Ubiquitina-Proteína Ligasas , Animales , Regulación hacia Abajo , Glucanos/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Ratones , Epilepsias Mioclónicas Progresivas , Enfermedades del Sistema Nervioso , Proteínas Tirosina Fosfatasas no Receptoras/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Biol Chem ; 296: 100150, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33277363

RESUMEN

Malstructured glycogen accumulates over time in Lafora disease (LD) and precipitates into Lafora bodies (LBs), leading to neurodegeneration and intractable fatal epilepsy. Constitutive reduction of glycogen synthase-1 (GYS1) activity prevents murine LD, but the effect of GYS1 reduction later in disease course is unknown. Our goal was to knock out Gys1 in laforin (Epm2a)-deficient LD mice after disease onset to determine whether LD can be halted in midcourse, or even reversed. We generated Epm2a-deficient LD mice with tamoxifen-inducible Cre-mediated Gys1 knockout. Tamoxifen was administered at 4 months and disease progression assessed at 12 months. We verified successful knockout at mRNA and protein levels using droplet digital PCR and Western blots. Glycogen determination and periodic acid-Schiff-diastase staining were used to analyze glycogen and LB accumulation. Immunohistochemistry using astrocytic (glial fibrillary acidic protein) and microglial (ionized calcium-binding adapter molecule 1) markers was performed to investigate neuroinflammation. In the disease-relevant organ, the brain, Gys1 mRNA levels were reduced by 85% and GYS1 protein depleted. Glycogen accumulation was halted at the 4-month level, while LB formation and neuroinflammation were significantly, though incompletely, prevented. Skeletal muscle analysis confirmed that Gys1 knockout inhibits glycogen and LB accumulation. However, tamoxifen-independent Cre recombination precluded determination of disease halting or reversal in this tissue. Our study shows that Gys1 knockdown is a powerful means to prevent LD progression, but this approach did not reduce brain glycogen or LBs to levels below those at the time of intervention. These data suggest that endogenous mechanisms to clear brain LBs are absent or, possibly, compromised in laforin-deficient murine LD.


Asunto(s)
Gliosis/prevención & control , Glucógeno Sintasa/fisiología , Inflamación/prevención & control , Enfermedad de Lafora/patología , Músculo Esquelético/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Animales , Femenino , Gliosis/metabolismo , Gliosis/patología , Inflamación/metabolismo , Inflamación/patología , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Tamoxifeno/administración & dosificación
7.
Brain ; 144(10): 2985-2993, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-33993268

RESUMEN

Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in mouse models of the disease. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analysed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.


Asunto(s)
Glucógeno Sintasa/administración & dosificación , Enfermedad de Lafora/tratamiento farmacológico , Enfermedad de Lafora/patología , Oligorribonucleótidos Antisentido/administración & dosificación , Animales , Femenino , Inyecciones Intraventriculares , Enfermedad de Lafora/genética , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética
8.
J Biol Chem ; 295(43): 14698-14709, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32817315

RESUMEN

The soluble α-polyglucan glycogen is a central metabolite enabling transient glucose storage to suit cellular energy needs. Glycogen storage diseases (GSDs) comprise over 15 entities caused by generalized or tissue-specific defects in enzymes of glycogen metabolism. In several, e.g. in Lafora disease caused by the absence of the glycogen phosphatase laforin or its interacting partner malin, degradation-resistant abnormally structured insoluble glycogen accumulates. Sensitive quantification methods for soluble and insoluble glycogen are critical to research, including therapeutic studies, in such diseases. This paper establishes methodological advancements relevant to glycogen metabolism investigations generally, and GSDs. Introducing a pre-extraction incubation method, we measure degradation-resistant glycogen in as little as 30 mg of skeletal muscle or a single hippocampus from Lafora disease mouse models. The digestion-resistant glycogen correlates with the disease-pathogenic insoluble glycogen and can readily be detected in very young mice where glycogen accumulation has just begun. Second, we establish a high-sensitivity glucose assay with detection of ATP depletion, enabling 1) quantification of α-glucans in cell culture using a medium-throughput assay suitable for assessment of candidate glycogen synthesis inhibitors, and 2) discovery of α-glucan material in healthy human cerebrospinal fluid, establishing a novel methodological platform for biomarker analyses in Lafora disease and other GSDs.


Asunto(s)
Glucanos/análisis , Glucanos/líquido cefalorraquídeo , Animales , Técnicas de Cultivo de Célula , Femenino , Enfermedad del Almacenamiento de Glucógeno/líquido cefalorraquídeo , Enfermedad del Almacenamiento de Glucógeno/patología , Células HEK293 , Hipocampo/patología , Humanos , Enfermedad de Lafora/líquido cefalorraquídeo , Enfermedad de Lafora/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología
9.
J Neurochem ; 157(6): 1897-1910, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32892347

RESUMEN

Mammalian glycogen chain lengths are subject to complex regulation, including by seven proteins (protein phosphatase-1 regulatory subunit 3, PPP1R3A through PPP1R3G) that target protein phosphatase-1 (PP1) to glycogen to activate the glycogen chain-elongating enzyme glycogen synthase and inactivate the chain-shortening glycogen phosphorylase. Lafora disease is a fatal neurodegenerative epilepsy caused by aggregates of long-chained, and as a result insoluble, glycogen, termed Lafora bodies (LBs). We previously eliminated PPP1R3C from a Lafora disease mouse model and studied the effect on LB formation. In the present work, we eliminate and study the effect of absent PPP1R3D. In the interim, brain cell type levels of all PPP1R3 genes have been published, and brain cell type localization of LBs clarified. Integrating these data we find that PPP1R3C is the major isoform in most tissues including brain. In the brain, PPP1R3C is expressed at 15-fold higher levels than PPP1R3D in astrocytes, the cell type where most LBs form. PPP1R3C deficiency eliminates ~90% of brain LBs. PPP1R3D is quantitatively a minor isoform, but possesses unique MAPK, CaMK2 and 14-3-3 binding domains and appears to have an important functional niche in murine neurons and cardiomyocytes. In neurons, it is expressed equally to PPP1R3C, and its deficiency eliminates ~50% of neuronal LBs. In heart, it is expressed at 25% of PPP1R3C where its deficiency eliminates ~90% of LBs. This work studies the role of a second (PPP1R3D) of seven PP1 subunits that regulate the structure of glycogen, toward better understanding of brain glycogen metabolism generally, and in Lafora disease.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Lafora/metabolismo , Miocardio/metabolismo , Neuronas/metabolismo , Proteína Fosfatasa 1/deficiencia , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Glucógeno/metabolismo , Humanos , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Neuronas/patología , Proteína Fosfatasa 1/genética
10.
PLoS Genet ; 14(4): e1007285, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29649218

RESUMEN

Epilepsy will affect nearly 3% of people at some point during their lifetime. Previous copy number variants (CNVs) studies of epilepsy have used array-based technology and were restricted to the detection of large or exonic events. In contrast, whole-genome sequencing (WGS) has the potential to more comprehensively profile CNVs but existing analytic methods suffer from limited accuracy. We show that this is in part due to the non-uniformity of read coverage, even after intra-sample normalization. To improve on this, we developed PopSV, an algorithm that uses multiple samples to control for technical variation and enables the robust detection of CNVs. Using WGS and PopSV, we performed a comprehensive characterization of CNVs in 198 individuals affected with epilepsy and 301 controls. For both large and small variants, we found an enrichment of rare exonic events in epilepsy patients, especially in genes with predicted loss-of-function intolerance. Notably, this genome-wide survey also revealed an enrichment of rare non-coding CNVs near previously known epilepsy genes. This enrichment was strongest for non-coding CNVs located within 100 Kbp of an epilepsy gene and in regions associated with changes in the gene expression, such as expression QTLs or DNase I hypersensitive sites. Finally, we report on 21 potentially damaging events that could be associated with known or new candidate epilepsy genes. Our results suggest that comprehensive sequence-based profiling of CNVs could help explain a larger fraction of epilepsy cases.


Asunto(s)
Variaciones en el Número de Copia de ADN , Epilepsia/genética , Estudios de Casos y Controles , Estudios de Cohortes , Humanos , Sitios de Carácter Cuantitativo , Secuenciación Completa del Genoma
11.
Epilepsia ; 61(7): e71-e78, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32645220

RESUMEN

Fibroblast growth-factor homologous factor (FHF1) gene variants have recently been associated with developmental and epileptic encephalopathy (DEE). FHF1 encodes a cytosolic protein that modulates neuronal sodium channel gating. We aim to refine the electroclinical phenotypic spectrum of patients with pathogenic FHF1 variants. We retrospectively collected clinical, genetic, neurophysiologic, and neuroimaging data of 17 patients with FHF1-DEE. Sixteen patients had recurrent heterozygous FHF1 missense variants: 14 had the recurrent p.Arg114His variant and two had a novel likely pathogenic variant p.Gly112Ser. The p.Arg114His variant is associated with an earlier onset and more severe phenotype. One patient carried a chromosomal microduplication involving FHF1. Twelve patients carried a de novo variant, five (29.5%) inherited from parents with gonadic or somatic mosaicism. Seizure onset was between 1 day and 41 months; in 76.5% it was within 30 days. Tonic seizures were the most frequent seizure type. Twelve patients (70.6%) had drug-resistant epilepsy, 14 (82.3%) intellectual disability, and 11 (64.7%) behavioral disturbances. Brain magnetic resonance imaging (MRI) showed mild cerebral and/or cerebellar atrophy in nine patients (52.9%). Overall, our findings expand and refine the clinical, EEG, and imaging phenotype of patients with FHF1-DEE, which is characterized by early onset epilepsy with tonic seizures, associated with moderate to severe ID and psychiatric features.


Asunto(s)
Encefalopatías/genética , Epilepsia/genética , Factores de Crecimiento de Fibroblastos/genética , Discapacidad Intelectual/genética , Fenotipo , Adolescente , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Encefalopatías/diagnóstico por imagen , Encefalopatías/fisiopatología , Niño , Preescolar , Electroencefalografía/métodos , Epilepsia/diagnóstico por imagen , Epilepsia/fisiopatología , Femenino , Humanos , Lactante , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/fisiopatología , Masculino , Estudios Retrospectivos , Adulto Joven
12.
Epidemiol Infect ; 148: e14, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32000877

RESUMEN

There is world-wide increasing interest in the consumption of unprocessed, natural food commodities including fresh (unpasteurised) milk and milk products. Consumers are actively seeking out raw milk, partly due to health reasons, but also for taste, freshness, closeness to the producer and to support local agriculture. The need for high levels of hygiene and safety in farms producing raw milk for direct consumption has long been recognised and has led to federal and industry-initiated systems for safe raw milk production. Raw milk producers in North America and Europe have demonstrated that raw milk, intended for direct consumption, can be produced safe and hygienic. The aim of this paper is to describe practices that have been developed for safe raw milk production. The German Vorzugsmilch is a federally regulated programme for legal raw milk production that was established already in the 1930s to provide raw milk with high hygienic standards controlled for zoonotic diseases to consumers. The Raw Milk Institute is a non-profit organisation established in California that has developed a voluntary safe raw milk programme in North America. RAWMI has developed a risk analysis and management system for raw milk dairy farmers to assist farmers in making individually tailored solutions for various production systems. In British Colombia, Canada, small herd share farms have employed good manufacturing practices, a risk management approach and performed monthly samples for pathogens and indicator bacteria to demonstrate safety and consistency. The major components of the raw milk systems applied, and the results of regular milk microbial indicator bacteria are presented. For the German system, the results from standard monthly pathogen tests are compared to zoonotic pathogen tests from other milk sources. The overall results indicate that raw milk can be produced with a high level of hygiene and safety in various systems.


Asunto(s)
Industria de Alimentos/métodos , Microbiología de Alimentos/métodos , Microbiología de Alimentos/normas , Inocuidad de los Alimentos/métodos , Leche/microbiología , Animales , Enfermedades Transmitidas por los Alimentos/prevención & control , Salud Global , Humanos
13.
Avian Pathol ; 49(4): 389-393, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32310002

RESUMEN

A clinical study was made into the abilities of nicarbazin and monensin and a nicarbazin + monensin combination to control Eimeria acervulina, E. maxima, and E. tenella in chickens. When included in the feed, at concentrations of 40 ppm nicarbazin or 40 ppm monensin, these products showed partial efficacy evaluated by daily weight gain (DWG) but no activity judged by daily feed intake (DFI) or feed conversion ratio (FCR). By contrast, the combination of 40 ppm nicarbazin + 40 ppm monensin provided complete control of infection judged by greater DWG and DFI, and lower FCR. Monensin at a concentration of 40 ppm was ineffective in preventing lesions caused by all three species. Nicarbazin at a concentration of 40 ppm was unable to suppress lesions of E. acervulina and E. maxima but was able to suppress lesions caused by E. tenella. Nicarbazin 40 ppm + monensin 40 ppm suppressed lesions of all three species. RESEARCH HIGHLIGHTS Nicarbazin or monensin at 40 ppm gave only partial control of Eimeria spp. A combination of 40 ppm nicarbazin + 40 ppm monensin controlled DWG, DFI and FCR. Nicarbazin or monensin at 40 ppm did not suppress all Eimeria spp. lesions. Nicarbazin 40 ppm + monensin 40 ppm suppressed lesions of all three species.


Asunto(s)
Coccidiosis/veterinaria , Coccidiostáticos/administración & dosificación , Eimeria/efectos de los fármacos , Monensina/administración & dosificación , Nicarbazina/administración & dosificación , Enfermedades de las Aves de Corral/tratamiento farmacológico , Animales , Coccidiosis/tratamiento farmacológico , Coccidiosis/parasitología , Sinergismo Farmacológico , Eimeria/genética , Masculino , Enfermedades de las Aves de Corral/virología
14.
Epilepsy Behav ; 103(Pt A): 106839, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932179

RESUMEN

Lafora disease (LD) is both a fatal childhood epilepsy and a glycogen storage disease caused by recessive mutations in either the Epilepsy progressive myoclonus 2A (EPM2A) or EPM2B genes. Hallmarks of LD are aberrant, cytoplasmic carbohydrate aggregates called Lafora bodies (LBs) that are a disease driver. The 5th International Lafora Epilepsy Workshop was recently held in Alcala de Henares, Spain. The workshop brought together nearly 100 clinicians, academic and industry scientists, trainees, National Institutes of Health (NIH) representation, and friends and family members of patients with LD. The workshop covered aspects of LD ranging from defining basic scientific mechanisms to elucidating a LD therapy or cure and a recently launched LD natural history study.


Asunto(s)
Congresos como Asunto/tendencias , Educación/tendencias , Internacionalidad , Enfermedad de Lafora/terapia , Animales , Humanos , Enfermedad de Lafora/epidemiología , Enfermedad de Lafora/genética , Mutación/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , España/epidemiología
15.
J Biol Chem ; 293(19): 7117-7125, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29483193

RESUMEN

Lafora disease (LD) is a fatal, autosomal recessive, glycogen-storage disorder that manifests as severe epilepsy. LD results from mutations in the gene encoding either the glycogen phosphatase laforin or the E3 ubiquitin ligase malin. Individuals with LD develop cytoplasmic, aberrant glycogen inclusions in nearly all tissues that more closely resemble plant starch than human glycogen. This Minireview discusses the unique window into glycogen metabolism that LD research offers. It also highlights recent discoveries, including that glycogen contains covalently bound phosphate and that neurons synthesize glycogen and express both glycogen synthase and glycogen phosphorylase.


Asunto(s)
Glucógeno/metabolismo , Enfermedad de Lafora/metabolismo , Neuronas/metabolismo , Animales , Conformación de Carbohidratos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Glucógeno/biosíntesis , Glucógeno/química , Glucógeno Fosforilasa/genética , Humanos , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Enfermedad de Lafora/terapia , Fosfatos/metabolismo , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras/genética , Ubiquitina-Proteína Ligasas/genética
16.
Am J Hum Genet ; 99(6): 1359-1367, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27866705

RESUMEN

Epileptic encephalopathies are a catastrophic group of epilepsies characterized by refractory seizures and cognitive arrest, often resulting from abnormal brain development. Here, we have identified an epileptic encephalopathy additionally featuring cerebral calcifications and coarse facial features caused by recessive loss-of-function mutations in DENND5A. DENND5A contains a DENN domain, an evolutionarily ancient enzymatic module conferring guanine nucleotide exchange factor (GEF) activity to multiple proteins serving as GEFs for Rabs, which are key regulators of membrane trafficking. DENND5A is detected predominantly in neuronal tissues, and its highest levels occur during development. Knockdown of DENND5A leads to striking alterations in neuronal development. Mechanistically, these changes appear to result from upregulation of neurotrophin receptors, leading to enhanced downstream signaling. Thus, we have identified a link between a DENN domain protein and neuronal development, dysfunction of which is responsible for a form of epileptic encephalopathy.


Asunto(s)
Encéfalo/patología , Epilepsia/genética , Mutación , Proteínas de Unión al GTP rab/genética , Adolescente , Animales , Niño , Consanguinidad , Femenino , Factores de Intercambio de Guanina Nucleótido , Humanos , Masculino , Neuronas/metabolismo , Células PC12 , Linaje , Ratas
17.
Epilepsia ; 60(9): 1881-1894, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31468518

RESUMEN

OBJECTIVE: Developmental epileptic encephalopathies (DEEs) are genetically heterogeneous severe childhood-onset epilepsies with developmental delay or cognitive deficits. In this study, we explored the pathogenic mechanisms of DEE-associated de novo mutations in the CACNA1A gene. METHODS: We studied the functional impact of four de novo DEE-associated CACNA1A mutations, including the previously described p.A713T variant and three novel variants (p.V1396M, p.G230V, and p.I1357S). Mutant cDNAs were expressed in HEK293 cells, and whole-cell voltage-clamp recordings were conducted to test the impacts on CaV 2.1 channel function. Channel localization and structure were assessed with immunofluorescence microscopy and three-dimensional (3D) modeling. RESULTS: We find that the G230V and I1357S mutations result in loss-of-function effects with reduced whole-cell current densities and decreased channel expression at the cell membrane. By contrast, the A713T and V1396M variants resulted in gain-of-function effects with increased whole-cell currents and facilitated current activation (hyperpolarized shift). The A713T variant also resulted in slower current decay. 3D modeling predicts conformational changes favoring channel opening for A713T and V1396M. SIGNIFICANCE: Our findings suggest that both gain-of-function and loss-of-function CACNA1A mutations are associated with similarly severe DEEs and that functional validation is required to clarify the underlying molecular mechanisms and to guide therapies.


Asunto(s)
Encefalopatías/genética , Canales de Calcio/genética , Mutación con Ganancia de Función , Síndrome de Lennox-Gastaut/genética , Mutación con Pérdida de Función , Espasmos Infantiles/genética , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Técnicas de Placa-Clamp , Fenotipo
18.
J Dairy Sci ; 102(7): 6391-6403, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31030920

RESUMEN

Antimicrobials are frequently administered to calves with diarrhea, despite evidence suggesting questionable efficacy. Even if efficacious, providing the appropriate therapy to an animal requires accurate disease detection. The objective of this study was to use previously collected data and compare clinical scoring by a veterinarian to treatment decisions by on-farm personnel. Data describing daily clinical scores and farm treatments were previously collected from 4 farms for calves from birth to age 28 d. In this data set, a total of 460 calves were enrolled. Daily observations and clinical assessments were made on each farm by the same veterinarian, for a total of 12,101 calf observation days. Farm personnel made all treatment decisions based on their own observations, and these treatments were recorded by study personnel. Overall, the cumulative incidence of a calf exhibiting at least one abnormal clinical sign over the 28-d observation period was 0.93, with cumulative incidences of 0.85 and 0.33 for diarrhea and dehydration, respectively. The cumulative incidence of any treatment (including antibiotics and electrolytes) was 0.85, although the majority of treatments used an antimicrobial. The farm-specific probabilities that a calf with clinical signs of dehydration or diarrhea, respectively, received fluid or electrolyte therapy ranged from 0.08 to 0.27 and 0.03 to 0.12. These probabilities were greater for the day a clinical sign was first observed. The farm-specific probabilities that a calf with clinical signs of diarrhea received an antimicrobial was 0.23 to 0.65, and the probability that a calf exhibiting clinical signs of respiratory disease received an antimicrobial was 0.33 to 0.76. The first observation of diarrhea had similar probabilities to those for all observations of diarrhea. There was greater probability of treatment for calves with their first observed abnormal respiratory signs. Probabilities that treatment with antimicrobials, or fluids or electrolytes, was associated with an abnormal clinical sign were low-that is, calves received treatments in the absence of any abnormal clinical signs. This study illustrates incongruity between treatment decisions by calf treaters (the designated personnel on each farm responsible for calf health assessment and treatment decisions) and those of an observer using a clinical scoring system to identify calves with abnormal clinical signs. These findings indicate opportunities and the need for dairy farmers and advisors to evaluate calf treatment protocols, reasons for treatment, and training programs for calf health and disease detection, as well as to develop monitoring programs for treatment protocol compliance and health outcomes following therapy.


Asunto(s)
Toma de Decisiones , Diarrea/veterinaria , Veterinarios/psicología , Animales , Antibacterianos/administración & dosificación , Bovinos , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/psicología , Diarrea/tratamiento farmacológico , Diarrea/epidemiología , Diarrea/psicología , Agricultores/psicología , Granjas/estadística & datos numéricos , Femenino , Humanos , Incidencia , Masculino , Embarazo , Estudios Retrospectivos
19.
Neuropediatrics ; 49(6): 357-362, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30336494

RESUMEN

Lafora's disease is a neurodegenerative disorder caused by recessive loss-of-function mutations in the EPM2A (laforin glycogen phosphatase) or EPM2B (malin E3 ubiquitin ligase) genes. Neuropathology is characterized by malformed precipitated glycogen aggregates termed Lafora bodies. Asymptomatic until adolescence, patients undergo first insidious then rapid progressive myoclonus epilepsy toward a vegetative state and death within a decade. Laforin and malin interact to regulate glycogen phosphorylation and chain length pattern, the latter critical to glycogen's solubility. Significant gaps remain in precise mechanistic understanding. However, demonstration that partial reduction in brain glycogen synthesis near-completely prevents the disease in its genetic animal models opens a direct present path to therapy.


Asunto(s)
Enfermedad de Lafora , Animales , Humanos , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Enfermedad de Lafora/fisiopatología , Enfermedad de Lafora/terapia
20.
Biochem J ; 474(20): 3403-3420, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28827282

RESUMEN

Glycogen storage disorders (GSDs) are caused by excessive accumulation of glycogen. Some GSDs [adult polyglucosan (PG) body disease (APBD), and Tarui and Lafora diseases] are caused by intracellular accumulation of insoluble inclusions, called PG bodies (PBs), which are chiefly composed of malconstructed glycogen. We developed an APBD patient skin fibroblast cell-based assay for PB identification, where the bodies are identified as amylase-resistant periodic acid-Schiff's-stained structures, and quantified. We screened the DIVERSet CL 10 084 compound library using this assay in high-throughput format and discovered 11 dose-dependent and 8 non-dose-dependent PB-reducing hits. Approximately 70% of the hits appear to act through reducing glycogen synthase (GS) activity, which can elongate glycogen chains and presumably promote PB generation. Some of these GS inhibiting hits were also computationally predicted to be similar to drugs interacting with the GS activator protein phosphatase 1. Our work paves the way to discovering medications for the treatment of PB-involving GSD, which are extremely severe or fatal disorders.


Asunto(s)
Fibroblastos/enzimología , Enfermedad del Almacenamiento de Glucógeno , Glucógeno Sintasa/metabolismo , Enfermedades del Sistema Nervioso , Adulto , Evaluación Preclínica de Medicamentos/métodos , Femenino , Enfermedad del Almacenamiento de Glucógeno/diagnóstico , Enfermedad del Almacenamiento de Glucógeno/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno/enzimología , Humanos , Masculino , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA