Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
JMIR Res Protoc ; 12: e46135, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37405822

ABSTRACT

BACKGROUND: The number of people with cognitive deficits and diseases, such as stroke, dementia, or attention-deficit/hyperactivity disorder, is rising due to an aging, or in the case of attention-deficit/hyperactivity disorder, a growing population. Neurofeedback training using brain-computer interfaces is emerging as a means of easy-to-use and noninvasive cognitive training and rehabilitation. A novel application of neurofeedback training using a P300-based brain-computer interface has previously shown potential to improve attention in healthy adults. OBJECTIVE: This study aims to accelerate attention training using iterative learning control to optimize the task difficulty in an adaptive P300 speller task. Furthermore, we hope to replicate the results of a previous study using a P300 speller for attention training, as a benchmark comparison. In addition, the effectiveness of personalizing the task difficulty during training will be compared to a nonpersonalized task difficulty adaptation. METHODS: In this single-blind, parallel, 3-arm randomized controlled trial, 45 healthy adults will be recruited and randomly assigned to the experimental group or 1 of 2 control groups. This study involves a single training session, where participants receive neurofeedback training through a P300 speller task. During this training, the task's difficulty is progressively increased, which makes it more difficult for the participants to maintain their performance. This encourages the participants to improve their focus. Task difficulty is either adapted based on the participants' performance (in the experimental group and control group 1) or chosen randomly (in control group 2). Changes in brain patterns before and after training will be analyzed to study the effectiveness of the different approaches. Participants will complete a random dot motion task before and after the training so that any transfer effects of the training to other cognitive tasks can be evaluated. Questionnaires will be used to estimate the participants' fatigue and compare the perceived workload of the training between groups. RESULTS: This study has been approved by the Maynooth University Ethics Committee (BSRESC-2022-2474456) and is registered on ClinicalTrials.gov (NCT05576649). Participant recruitment and data collection began in October 2022, and we expect to publish the results in 2023. CONCLUSIONS: This study aims to accelerate attention training using iterative learning control in an adaptive P300 speller task, making it a more attractive training option for individuals with cognitive deficits due to its ease of use and speed. The successful replication of the results from the previous study, which used a P300 speller for attention training, would provide further evidence to support the effectiveness of this training tool. TRIAL REGISTRATION: ClinicalTrials.gov NCT05576649; https://clinicaltrials.gov/ct2/show/NCT05576649. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/46135.

2.
Brain ; 142(3): 542-559, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30668673

ABSTRACT

Biallelic pathogenic variants in PLPBP (formerly called PROSC) have recently been shown to cause a novel form of vitamin B6-dependent epilepsy, the pathophysiological basis of which is poorly understood. When left untreated, the disease can progress to status epilepticus and death in infancy. Here we present 12 previously undescribed patients and six novel pathogenic variants in PLPBP. Suspected clinical diagnoses prior to identification of PLPBP variants included mitochondrial encephalopathy (two patients), folinic acid-responsive epilepsy (one patient) and a movement disorder compatible with AADC deficiency (one patient). The encoded protein, PLPHP is believed to be crucial for B6 homeostasis. We modelled the pathogenicity of the variants and developed a clinical severity scoring system. The most severe phenotypes were associated with variants leading to loss of function of PLPBP or significantly affecting protein stability/PLP-binding. To explore the pathophysiology of this disease further, we developed the first zebrafish model of PLPHP deficiency using CRISPR/Cas9. Our model recapitulates the disease, with plpbp-/- larvae showing behavioural, biochemical, and electrophysiological signs of seizure activity by 10 days post-fertilization and early death by 16 days post-fertilization. Treatment with pyridoxine significantly improved the epileptic phenotype and extended lifespan in plpbp-/- animals. Larvae had disruptions in amino acid metabolism as well as GABA and catecholamine biosynthesis, indicating impairment of PLP-dependent enzymatic activities. Using mass spectrometry, we observed significant B6 vitamer level changes in plpbp-/- zebrafish, patient fibroblasts and PLPHP-deficient HEK293 cells. Additional studies in human cells and yeast provide the first empirical evidence that PLPHP is localized in mitochondria and may play a role in mitochondrial metabolism. These models provide new insights into disease mechanisms and can serve as a platform for drug discovery.


Subject(s)
Epilepsy/etiology , Proteins/genetics , Proteins/metabolism , Animals , Disease Models, Animal , Epilepsy/physiopathology , Female , HEK293 Cells , Humans , Male , Phenotype , Pyridoxal Phosphate/therapeutic use , Pyridoxine/deficiency , Vitamin B 6/metabolism , Vitamin B 6 Deficiency/genetics , Vitamin B 6 Deficiency/metabolism , Zebrafish
3.
Mar Biotechnol (NY) ; 19(6): 563-569, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28980193

ABSTRACT

As the world increasingly relies on aquaculture operations to meet rising seafood demands, reliable biocontainment measures for farmed fish stocks are desired to minimize ecological impacts arising from interactions of cultured fish with wild populations. One possible biocontainment strategy is to induce a dietary dependence on a vitamin, such as thiamine (vitamin B1), required for survival. Fish expressing thiaminase (an enzyme that degrades thiamine) within a confined aquaculture facility could receive supplemental thiamine to allow survival and normal growth, whereas escapees lacking this dietary rescue would die from thiamine deficiency. To test the concept and efficacy of such a dietary dependency system (for potential future use in larger aquaculture species), we expressed thiaminase in zebrafish as a test model. We drove the expression of thiaminase under the strong ubiquitous and constitutive control of the CMV promoter which resulted in non-viable fish, indicating that the thiaminase sequence kills fish. However, the CMV promoter is too strong to allow conditional survival since the lethality could not be rescued by exogenous thiamine provided as a supplement to typical food. In addition, microinjection of 0.5 pg of thiaminase mRNA in zebrafish embryos at the one-cell stage resulted in 50% larval mortality at 5 days post-fertilization (dpf), which was partially rescued by thiamine supplementation. Evaluating the efficacy of biocontainment strategies helps assess which methods can reliably prevent ecological impacts arising from breaches in physical containment systems that release engineered organisms to nature, and consequently provides critical information for use in regulatory risk assessment processes.


Subject(s)
Hydrolases/genetics , Thiamine Deficiency/veterinary , Zebrafish/genetics , Animals , Animals, Genetically Modified , Aquaculture/methods , Bacillus thuringiensis/genetics , Diet/veterinary , Embryo, Nonmammalian/metabolism , Hydrolases/metabolism , Introduced Species , RNA, Messenger/administration & dosage , Thiamine/administration & dosage , Thiamine Deficiency/mortality , Zebrafish/embryology , Zebrafish/metabolism
4.
Aquat Toxicol ; 175: 73-80, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26994370

ABSTRACT

Methylmercury (MeHg) is a known neurotoxicant affecting the central nervous system but effects on dopaminergic (DA) neurons are not well understood. Wild-type zebrafish (Danio rerio) and two transgenic lines: Tg(dat:eGFP) expressing enhanced green fluorescent protein (eGFP) in DA neuron clusters and Tg(dat:tom20 MLS-mCherry) expressing red fluorescence (mCherry) targeted to mitochondria of DA neurons were used to evaluate the effects of micromolar MeHg exposure on DA neuron and whole animal motor function during early development. Three-day-old larvae were exposed to micromolar concentrations of MeHg (0.03, 0.06, and 0.3µM) in system water. Exposure to 0.3µM MeHg caused mortality and significant morphological abnormalities including edema, curvature of the spine, and hemorrhages in zebrafish larvae after a 48h exposure period. At 0.06µM MeHg, the appearance of morphological abnormalities was delayed for 72h and far less severe, whereas 0.03µM MeHg did not cause any morphological defects or mortalities. A delayed but significant reduction in locomotor ability and mCherry fluorescence in specific brain regions in the 0.06µM MeHg exposed larvae suggests that DA neuron function rather than neuron numbers was compromised. Double immunolabeling with tyrosine hydroxylase and pan neural staining showed no effect of MeHg exposure. We have established Tg(dat:tom20 MLS-mCherry) zebrafish larvae as a model which can be used to assess MeHg neurotoxicity and that exposure to low dose MeHg (0.06µM) during development may predispose DA neurons to impairment caused by changes in mitochondrial dynamics.


Subject(s)
Dopaminergic Neurons/drug effects , Larva/drug effects , Methylmercury Compounds/toxicity , Mitochondria/drug effects , Water Pollutants, Chemical/toxicity , Zebrafish/physiology , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Swimming
5.
Zebrafish ; 12(5): 349-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26355474

ABSTRACT

Genetic mutations and environmental toxins are known to affect mitochondrial health and have been implicated in the progressive degeneration of dopaminergic neurons in Parkinson's disease. To visualize mitochondria in dopaminergic neurons of live zebrafish, we used the regulatory elements of the dopamine transporter (dat) gene to target a reporter, mCherry, after fusion with the mitochondrial localizing signal (MLS) of Tom20. Immunoblot analysis of mitochondrial and cytosolic fractions from Tg(dat:tom20 MLS-mCherry) larvae shows that mCherry is efficiently targeted to the mitochondria. Confocal imaging of live fish was carried out from 1 day postfertilization (dpf) to 9 dpf. We also colocalized dat mRNA expression with the mCherry protein in the olfactory bulb (OB), subpallium (SP), pretectum (Pr), diencephalic clusters 2 and 3 (DC2/3), caudal hypothalamus (Hc), locus coeruleus (LC), anterior preoptic area (POa), retinal amacrine cells (RAC), caudal hypothalamus (Hc), and preoptic area (PO). Treating Tg(dat:tom20 MLS-mCherry) larvae with the dopaminergic neurotoxin MPTP (1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine) at 2 or 3 dpf resulted in a decrease in mCherry fluorescence in the pretectum, olfactory bulb, subpallium, diencephalic clusters 2 and 3, and the caudal hypothalamus. Labeling of mitochondria in nigrostriatal dopaminergic neurons of zebrafish could allow their visualization in vivo following genetic or pharmacological manipulations.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/metabolism , Mitochondria/drug effects , Neurotoxins/pharmacology , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Fluorescence , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Organ Specificity , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Red Fluorescent Protein
6.
J Neurochem ; 135(2): 249-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26118896

ABSTRACT

To determine the impact of a controlled loss of dopaminergic neurons on locomotor function, we generated transgenic zebrafish, Tg(dat:CFP-NTR), expressing a cyan fluorescent protein-nitroreductase fusion protein (CFP-NTR) under the control of dopamine transporter (dat) cis-regulatory elements. Embryonic and larval zebrafish express the transgene in several groups of dopaminergic neurons, notably in the olfactory bulb, telencephalon, diencephalon and caudal hypothalamus. Administration of the pro-drug metronidazole (Mtz) resulted in activation of caspase 3 in CFP-positive neurons and in a reduction in dat-positive cells by 5 days post-fertilization (dpf). Loss of neurons coincided with impairments in global locomotor parameters such as swimming distance, percentage of time spent moving, as well as changes in tail bend parameters such as time to maximal bend and angular velocity. Dopamine levels were transiently decreased following Mtz administration. Recovery of some of the locomotor parameters was observed by 7 dpf. However, the total numbers of dat-expressing neurons were still decreased at 7, 12, or 14 dpf, even though there was evidence for production of new dat-expressing cells. Tg(dat:CFP-NTR) zebrafish provide a model to correlate altered dopaminergic neuron numbers with locomotor function and to investigate factors influencing regeneration of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/drug effects , Dyskinesia, Drug-Induced/genetics , Locomotion/drug effects , Animals , Animals, Genetically Modified , Brain/cytology , Brain/growth & development , Dyskinesia, Drug-Induced/physiopathology , Green Fluorescent Proteins , Humans , Larva/growth & development , MPTP Poisoning/pathology , Metronidazole/pharmacology , Nerve Regeneration , Swimming , Zebrafish
7.
Dev Biol ; 373(2): 258-66, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23206891

ABSTRACT

Spontaneous intracranial hemorrhage is a debilitating form of stroke, often leading to death or permanent cognitive impairment. Many of the causative genes and the underlying mechanisms implicated in developmental cerebral-vascular malformations are unknown. Recent in vitro and in vivo studies in mice have shown inhibition of the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) pathway to be effective in stabilizing cranial vessels. Using a combination of pharmacological and genetic approaches to specifically inhibit the HMGCR pathway in zebrafish (Danio rerio), we demonstrate a requirement for this metabolic pathway in developmental vascular stability. Here we report that inhibition of HMGCR function perturbs cerebral-vascular stability, resulting in progressive dilation of blood vessels, followed by vessel rupture, mimicking cerebral cavernous malformation (CCM)-like lesions in humans and murine models. The hemorrhages in the brain are rescued by prior exogenous supplementation with geranylgeranyl pyrophosphate (GGPP), a 20-carbon metabolite of the HMGCR pathway, required for the membrane localization and activation of Rho GTPases. Consistent with this observation, morpholino-induced depletion of the ß-subunit of geranylgeranyltransferase I (GGTase I), an enzyme that facilitates the post-translational transfer of the GGPP moiety to the C-terminus of Rho family of GTPases, mimics the cerebral hemorrhaging induced by the pharmacological and genetic ablation of HMGCR. In embryos with cerebral hemorrhage, the endothelial-specific expression of cdc42, a Rho GTPase involved in the regulation of vascular permeability, was significantly reduced. Taken together, our data reveal a metabolic contribution to the stabilization of nascent cranial vessels, requiring protein geranylgeranylation acting downstream of the HMGCR pathway.


Subject(s)
Cerebrum/blood supply , Cerebrum/embryology , Hydroxymethylglutaryl CoA Reductases/metabolism , Prenylation , Signal Transduction , Zebrafish/embryology , Alkyl and Aryl Transferases/metabolism , Animals , Atorvastatin , Cerebral Hemorrhage/embryology , Cerebral Hemorrhage/pathology , Cerebrum/drug effects , Cerebrum/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/pathology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Morpholinos/pharmacology , Polyisoprenyl Phosphates/biosynthesis , Prenylation/drug effects , Pyrroles/pharmacology , Signal Transduction/drug effects , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
J Neurochem ; 122(1): 196-207, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22506991

ABSTRACT

Many genes associated with familial Parkinson's disease contribute to mitochondrial morphology and function. Some of these genes, for example, Pink1 and Parkin, are part of a common pathway. The presenilin-associated rhomboid-like (PARL) gene was recently linked to familial Parkinson's disease. The PARL gene product is found in the inner mitochondrial membrane and cleaves the optic atrophy 1 protein, involved in mitochondrial morphology and apoptosis. In Drosophila, the PARL-related rhomboid-7 gene acts upstream of pink1 and parkin. However, such a genetic relationship is still unknown in vertebrates. Here, we show that the zebrafish genome comprises two parl paralogs: parla and parlb. Morpholino-mediated loss of parla and/or parlb function resulted in mild neurodegeneration, as evidenced by a lower density of dopaminergic neurons. Patterning of dopaminergic neurons was also perturbed in the ventral diencephalon. Morphants exhibited extensive cell death throughout the entire body as well as increased larval mortality. The morphant phenotype could be rescued by injection of human PARL mRNA, but not catalytically inactive PARL, suggesting functional conservation between the human and zebrafish proteins. More importantly, the zebrafish pink1 mRNA as well as the human PINK1 mRNA, but not kinase-dead nor Parkinson's disease-linked mutant PINK1 mRNA, also rescued the morphant phenotype, providing evidence that Parl genes may function upstream of Pink1, as part of a conserved pathway in vertebrates.


Subject(s)
Body Patterning/genetics , Dopaminergic Neurons/physiology , Metalloproteases/genetics , Mitochondrial Proteins/deficiency , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Acridine Orange , Animals , Animals, Genetically Modified , Cell Death/drug effects , Cell Death/genetics , Cloning, Molecular , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Larva/cytology , Mitochondrial Proteins/genetics , Morpholinos/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Messenger/metabolism , Sequence Alignment/methods , Tyrosine 3-Monooxygenase/metabolism , Zebrafish
9.
Curr Neurol Neurosci Rep ; 11(3): 274-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21271309

ABSTRACT

The zebrafish, Danio rerio, has been established as an excellent vertebrate model for the study of developmental biology and gene function. It also has proven to be a valuable model to study human diseases. Here, we reviewed recent publications using zebrafish to study the pathology of human neurodegenerative diseases including Parkinson's, Huntington's, and Alzheimer's. These studies indicate that zebrafish genes and their human homologues have conserved functions with respect to the etiology of neurodegenerative diseases. The characteristics of the zebrafish and the experimental approaches to which it is amenable make this species a useful complement to other animal models for the study of pathologic mechanisms of neurodegenerative diseases and for the screening of compounds with therapeutic potential.


Subject(s)
Disease Models, Animal , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Zebrafish/genetics , Alzheimer Disease/genetics , Animals , Humans , Huntington Disease/genetics , Intracellular Signaling Peptides and Proteins/genetics , LDL-Receptor Related Proteins/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Membrane Transport Proteins/genetics , Oncogene Proteins/genetics , Parkinson Disease/genetics , Protein Deglycase DJ-1 , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , tau Proteins/genetics
10.
Eur J Neurosci ; 31(4): 623-33, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20141529

ABSTRACT

Mutations in the human PTEN-induced kinase 1 (PINK1) gene are linked to recessive familial Parkinson's disease. Animal models of altered PINK1 function vary greatly in their phenotypic characteristics. Drosophila pink1 mutants exhibit mild dopaminergic neuron degeneration and locomotion defects. Such defects are not observed in mice with targeted null mutations in pink1, although these mice exhibit impaired dopamine release and synaptic plasticity. Here, we report that in zebrafish, morpholino-mediated knockdown of pink1 function did not cause large alterations in the number of dopaminergic neurons in the ventral diencephalon. However, the patterning of these neurons and their projections are perturbed. This is accompanied by locomotor dysfunction, notably impaired response to tactile stimuli and reduced swimming behaviour. All these defects can be rescued by expression of an exogenous pink1 that is not a target of the morpholinos used. These results indicate that normal PINK1 function during development is necessary for the proper positioning of populations of dopaminergic neurons and for the establishment of neuronal circuits in which they are implicated.


Subject(s)
Diencephalon/growth & development , Protein Kinases/genetics , Swimming/physiology , Touch Perception/physiology , Zebrafish , Amino Acid Sequence , Animals , Diencephalon/anatomy & histology , Diencephalon/drug effects , Diencephalon/metabolism , Dopamine/metabolism , Larva/drug effects , Larva/physiology , Molecular Sequence Data , Neurons/metabolism , Neurons/physiology , Oligonucleotides, Antisense/pharmacology , Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...