Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 15(6): 429, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890273

RESUMEN

Tauopathies are characterised by the pathological accumulation of misfolded tau. The emerging view is that toxic tau species drive synaptic dysfunction and potentially tau propagation before measurable neurodegeneration is evident, but the underlying molecular events are not well defined. Human non-mutated 0N4R tau (tauWT) and P301L mutant 0N4R tau (tauP301L) were expressed in mouse primary cortical neurons using adeno-associated viruses to monitor early molecular changes and synaptic function before the onset of neuronal loss. In this model tauP301L was differentially phosphorylated relative to tauwt with a notable increase in phosphorylation at ser262. Affinity purification - mass spectrometry combined with tandem mass tagging was used to quantitatively compare the tauWT and tauP301L interactomes. This revealed an enrichment of tauP301L with ribosomal proteins but a decreased interaction with the proteasome core complex and reduced tauP301L degradation. Differences in the interaction of tauP301L with members of a key synaptic calcium-calmodulin signalling pathway were also identified, most notably, increased association with CaMKII but reduced association with calcineurin and the candidate AD biomarker neurogranin. Decreased association of neurogranin to tauP301L corresponded with the appearance of enhanced levels of extracellular neurogranin suggestive of potential release or leakage from synapses. Finally, analysis of neuronal network activity using micro-electrode arrays showed that overexpression of tauP301L promoted basal hyperexcitability coincident with these changes in the tau interactome and implicating tau in specific early alterations in synaptic function.


Asunto(s)
Neuronas , Proteínas tau , Animales , Proteínas tau/metabolismo , Proteínas tau/genética , Humanos , Ratones , Neuronas/metabolismo , Fosforilación , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/genética , Sinapsis/metabolismo , Neurogranina/metabolismo , Neurogranina/genética , Calcineurina/metabolismo
2.
Sci Rep ; 12(1): 15847, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36151233

RESUMEN

Alzheimer's disease (AD) involves pathological processing of amyloid precursor protein (APP) into amyloid-ß and microtubule associated protein Tau (MAPT) into hyperphosphorylated Tau tangles leading to neurodegeneration. Only 5% of AD cases are familial making it difficult to predict who will develop the disease thereby hindering our ability to treat the causes of the disease. A large population who almost certainly will, are those with Down syndrome (DS), who have a 90% lifetime incidence of AD. DS is caused by trisomy of chromosome 21 resulting in three copies of APP and other AD-associated genes, like dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) overexpression. This implies that DYRK1a inhibitors may have therapeutic potential for DS and AD, however It is not clear how overexpression of each of these genes contributes to the pathology of each disease as well as how effective a DYRK1A inhibitor would be at suppressing any of these. To address this knowledge gap, we used Drosophila models with human Tau, human amyloid-ß or fly DYRK1A (minibrain (mnb)) neuronal overexpression resulting in photoreceptor neuron degeneration, premature death, decreased locomotion, sleep and memory loss. DYRK1A small molecule Type 1 kinase inhibitors (DYR219 and DYR533) were effective at suppressing these disease relevant phenotypes confirming their therapeutic potential.


Asunto(s)
Enfermedad de Alzheimer , Síndrome de Down , Síndromes de Neurotoxicidad , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/genética , Síndrome de Down/metabolismo , Drosophila , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tirosina/metabolismo , Proteínas tau/metabolismo , Quinasas DyrK
3.
Clin Genet ; 102(6): 494-502, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36046955

RESUMEN

Cerebral palsy (CP) causes neurological disability in early childhood. Hypoxic-ischaemic injury plays a major role in its aetiology, nevertheless, genetic and epigenetic factors may contribute to the clinical presentation. Mutations in ADD3 (encoding γ-adducin) gene have been described in a monogenic form of spastic quadriplegic cerebral palsy (OMIM 601568). We studied a 16-year-old male with spastic diplegia. Several investigations including neurometabolic testing, brain and spine magnetic resonance imaging (MRI) and CGH-Array were normal. Further, clinical genetics assessment and whole exome sequencing (WES) gave the diagnosis. We generated an animal model using Drosophila to study the effects of γ-adducin loss and gain of function. WES revealed a biallelic variant in the ADD3 gene, NM_016824.5(ADD3): c.1100G > A, p.(Gly367Asp). Mutations in this gene have been described as an ultra-rare autosomal recessive, which is a known form of inherited cerebral palsy. Molecular modelling suggests that this mutation leads to a loss of structural integrity of γ-adducin and is therefore expected to result in a decreased level of functional protein. Pan-neuronal over-expression or knock-down of the Drosophila ortholog of ADD3 called hts caused a reduction of life span and impaired locomotion thereby phenocopying aspects of the human disease. Our animal experiments present a starting point to understand the biological processes underpinning the clinical phenotype and pathogenic mechanisms, to gain insights into potential future methods for treating or preventing ADD3 related spastic quadriplegic cerebral palsy.


Asunto(s)
Parálisis Cerebral , Paraparesia Espástica , Paraplejía Espástica Hereditaria , Animales , Masculino , Preescolar , Humanos , Adolescente , Drosophila/genética , Paraparesia Espástica/genética , Espasticidad Muscular , Mutación , Paraplejía Espástica Hereditaria/genética , Proteínas de Unión a Calmodulina/genética
4.
Front Pharmacol ; 13: 881385, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35928283

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease which is becoming increasingly prevalent due to ageing populations resulting in huge social, economic, and health costs to the community. Despite the pathological processing of genes such as Amyloid Precursor Protein (APP) into Amyloid-ß and Microtubule Associated Protein Tau (MAPT) gene, into hyperphosphorylated Tau tangles being known for decades, there remains no treatments to halt disease progression. One population with increased risk of AD are people with Down syndrome (DS), who have a 90% lifetime incidence of AD, due to trisomy of human chromosome 21 (HSA21) resulting in three copies of APP and other AD-associated genes, such as DYRK1A (Dual specificity tyrosine-phosphorylation-regulated kinase 1A) overexpression. This suggests that blocking DYRK1A might have therapeutic potential. However, it is still not clear to what extent DYRK1A overexpression by itself leads to AD-like phenotypes and how these compare to Tau and Amyloid-ß mediated pathology. Likewise, it is still not known how effective a DYRK1A antagonist may be at preventing or improving any Tau, Amyloid-ß and DYRK1a mediated phenotype. To address these outstanding questions, we characterised Drosophila models with targeted overexpression of human Tau, human Amyloid-ß or the fly orthologue of DYRK1A, called minibrain (mnb). We found targeted overexpression of these AD-associated genes caused degeneration of photoreceptor neurons, shortened lifespan, as well as causing loss of locomotor performance, sleep, and memory. Treatment with the experimental DYRK1A inhibitor PST-001 decreased pathological phosphorylation of human Tau [at serine (S) 262]. PST-001 reduced degeneration caused by human Tau, Amyloid-ß or mnb lengthening lifespan as well as improving locomotion, sleep and memory loss caused by expression of these AD and DS genes. This demonstrated PST-001 effectiveness as a potential new therapeutic targeting AD and DS pathology.

5.
Neurobiol Dis ; 170: 105752, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35569721

RESUMEN

Alzheimer's disease (AD) is the most prevalent neurodegenerative disease placing a great burden on people living with it, carers and society. Yet, the underlying patho-mechanisms remain unknown and treatments limited. To better understand the molecular changes associated with AD, genome-wide association studies (GWAS) have identified hundreds of candidate genes linked to the disease, like the receptor tyrosine kinase EphA1. However, demonstration of whether and how these genes cause pathology is largely lacking. Here, utilising fly genetics, we generated the first Drosophila model of human wild-type and P460L mutant EphA1 and tested the effects of Eph/ephrin signalling on AD-relevant behaviour and neurophysiology. We show that EphA1 mis-expression did not cause neurodegeneration, shorten lifespan or affect memory but flies mis-expressing the wild-type or mutant receptor were hyper-aroused, had reduced sleep, a stronger circadian rhythm and increased clock neuron activity and excitability. Over-expression of endogenous fly Eph and RNAi-mediated knock-down of Eph and its ligand ephrin affected sleep architecture and neurophysiology. Eph over-expression led to stronger circadian morning anticipation while ephrin knock-down impaired memory. A dominant negative form of the GTPase Rho1, a potential intracellular effector of Eph, led to hyper-aroused flies, memory impairment, less anticipatory behaviour and neurophysiological changes. Our results demonstrate a role of Eph/ephrin signalling in a range of behaviours affected in AD. This presents a starting point for studies into the underlying mechanisms of AD including interactions with other AD-associated genes, like Rho1, Ankyrin, Tau and APP with the potential to identify new targets for treatment.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedad de Alzheimer/genética , Animales , Drosophila , Efrinas/genética , Estudio de Asociación del Genoma Completo , Humanos , Neurofisiología , Receptores de la Familia Eph/genética
6.
IBRO Neurosci Rep ; 10: 196-207, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34041508

RESUMEN

Botulinum neurotoxins (BoNTs) are extremely potent naturally occurring poisons that act by silencing neurotransmission. Intriguingly, in addition to preventing presynaptic vesicle fusion, BoNT serotype A (BoNT/A) can also promote axonal regeneration in preclinical models. Here we report that the non-toxic C-terminal region of the receptor-binding domain of heavy chain BoNT/A (HCC/A) activates the small GTPase Rac1 and ERK pathway to potentiate axonal outgrowth, dendritic protrusion formation and synaptic vesicle release in hippocampal neurons. These data are consistent with HCC/A exerting neurotrophic properties, at least in part, independent of any BoNT catalytic activity or toxic effect.

7.
Sci Rep ; 11(1): 2061, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479461

RESUMEN

Globally, neonicotinoids are the most used insecticides, despite their well-documented sub-lethal effects on beneficial insects. Neonicotinoids are nicotinic acetylcholine receptor agonists. Memory, circadian rhythmicity and sleep are essential for efficient foraging and pollination and require nicotinic acetylcholine receptor signalling. The effect of field-relevant concentrations of the European Union-banned neonicotinoids: imidacloprid, clothianidin, thiamethoxam and thiacloprid were tested on Drosophila memory, circadian rhythms and sleep. Field-relevant concentrations of imidacloprid, clothianidin and thiamethoxam disrupted learning, behavioural rhythmicity and sleep whilst thiacloprid exposure only affected sleep. Exposure to imidacloprid and clothianidin prevented the day/night remodelling and accumulation of pigment dispersing factor (PDF) neuropeptide in the dorsal terminals of clock neurons. Knockdown of the neonicotinoid susceptible Dα1 and Dß2 nicotinic acetylcholine receptor subunits in the mushroom bodies or clock neurons recapitulated the neonicotinoid like deficits in memory or sleep/circadian behaviour respectively. Disruption of learning, circadian rhythmicity and sleep are likely to have far-reaching detrimental effects on beneficial insects in the field.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Memoria/efectos de los fármacos , Receptores Nicotínicos/genética , Sueño/efectos de los fármacos , Animales , Ritmo Circadiano/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Guanidinas/farmacología , Insecticidas/efectos adversos , Insecticidas/farmacología , Memoria/fisiología , Neonicotinoides/efectos adversos , Neonicotinoides/farmacología , Neuropéptidos/genética , Agonistas Nicotínicos/farmacología , Nitrocompuestos/farmacología , Sueño/genética , Tiametoxam/farmacología , Tiazinas/farmacología , Tiazoles/farmacología
8.
Epilepsia ; 61(4): 810-821, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32112430

RESUMEN

OBJECTIVE: Genetic variants in STXBP1, which encodes the conserved exocytosis protein Munc18-1, are associated with a variety of infantile epilepsy syndromes. We aimed to develop an in vivo Caenorhabditis elegans model that could be used to test the pathogenicity of such variants in a cost-effective manner. METHODS: The CRISPR/Cas9 method was used to introduce a null mutation into the unc-18 gene (the C. elegans orthologue of STXBP1), thereby creating a paralyzed worm strain. We subsequently rescued this strain with transgenes encoding the human STXBP1/Munc18-1 protein (wild-type and eight different epilepsy-associated missense variants). The resulting humanized worm strains were then analyzed via behavioral, electrophysiological, and biochemical approaches. RESULTS: Transgenic expression of wild-type human STXBP1 protein fully rescued locomotion in both solid and liquid media to the same level as the standard wild-type worm strain, Bristol N2. Six variant strains (E59K, V84D, C180Y, R292H, L341P, R551C) exhibited impaired locomotion, whereas two (P335L, R406H) were no different from worms expressing wild-type STXBP1. Electrophysiological recordings revealed that all eight variant strains displayed less frequent and more irregular pharyngeal pumping in comparison to wild-type STXBP1-expressing strains. Four strains (V84D, C180Y, R292H, P335L) exhibited pentylenetetrazol-induced convulsions in an acute assay of seizure-like activity, in contrast to worms expressing wild-type STXBP1. No differences were seen between wild-type and variant STXBP1 strains in terms of mRNA abundance. However, STXBP1 protein levels were reduced to 20%-30% of wild-type in all variants, suggesting that the mutations result in STXBP1 protein instability. SIGNIFICANCE: The approach described here is a cost-effective in vivo method for establishing the pathogenicity of genetic variants in STXBP1 and potentially other conserved neuronal proteins. Furthermore, the humanized strains we created could potentially be used in the future for high-throughput drug screens to identify novel therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/genética , Proteínas Munc18/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Humanos , Mutación , Proteínas de Transporte Vesicular/genética
9.
Stem Cells Int ; 2019: 8907570, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809265

RESUMEN

Smooth muscle cell- (SMC-) based tissue engineering provides a promising therapeutic strategy for SMC-related disorders. It has been demonstrated that human dental pulp stem cells (DPSCs) possess the potential to differentiate into mature bladder SMCs by induction with condition medium (CM) from bladder SMC culture, in combination with the transforming growth factor-ß1 (TGF-ß1). However, the molecular mechanism of SMC differentiation from DPSCs has not been fully uncovered. The canonical Wnt signaling (also known as Wnt/ß-catenin) pathway plays an essential role in stem cell fate decision. The aim of this study is to explore the regulation via GSK3ß and associated downstream effectors for SMC differentiation from DPSCs. We characterized one of our DPSC clones with the best proliferation and differentiation abilities. This stem cell clone has shown the capacity to generate a smooth muscle layer-like phenotype after an extended differentiation duration using the SMC induction protocol we established before. We further found that Wnt-GSK3ß/ß-catenin signaling is involved in the process of SMC differentiation from DPSCs, as well as a serial of growth factors, including TGF-ß1, basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), hepatocyte growth factor (HGF), platelet-derived growth factor-homodimer polypeptide of B chain (BB) (PDGF-BB), and vascular endothelial growth factor (VEGF). Pharmacological inhibition on the canonical Wnt-GSK3ß/ß-catenin pathway significantly downregulated GSK3ß phosphorylation and ß-catenin activation, which in consequence reduced the augmented expression of the growth factors (including TGF-ß1, HGF, PDGF-BB, and VEGF) as well as SMC markers (especially myosin) at a late stage of SMC differentiation. These results suggest that the canonical Wnt-GSK3ß/ß-catenin pathway contributes to DPSC differentiation into mature SMCs through the coordination of different growth factors.

10.
Stem Cell Res ; 33: 69-78, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30321831

RESUMEN

An essential aspect of stem cell in vitro culture and in vivo therapy is achieving sustained levels of growth factors to support stem cell survival and expansion, while maintaining their multipotency and differentiation potential. This study investigated the ability of dextrin (~74,000 g/mol; 27.8 mol% succinoylation) conjugated to epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF; or FGF-2) (3.9 and 6.7% w/w protein loading, respectively) to support the expansion and differentiation of stem cells in vitro via sustained, controllable growth factor release. Supplementation of mouse neural stem cells (mNSCs) with dextrin-growth factor conjugates led to greater and prolonged proliferation compared to unbound EGF/bFGF controls, with no detectable apoptosis after 7 days of treatment. Immunocytochemical detection of neural precursor (nestin) and differentiation (Olig2, MAP2, GFAP) markers verified that controlled release of dextrin-conjugated growth factors preserves stem cell properties of mNSCs for up to 7 days. These results show the potential of dextrin-growth factor conjugates for localized delivery of bioactive therapeutic agents to support stem cell expansion and differentiation, and as an adjunct to direct neuronal repair.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Dextrinas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células-Madre Neurales/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos
11.
Biochem Biophys Res Commun ; 500(3): 645-649, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29678571

RESUMEN

Neural stem cells (NSCs) are self-renewing multipotent stem cells that can be proliferated in vitro and differentiated into neuronal and/or glial lineages, making them an ideal model to study the processes involved in neuronal differentiation. Here we have used NSCs to investigate the role of the transcription factor MEF2A in neuronal differentiation and development in vitro. We show that although MEF2A is present in undifferentiated NSCs, following differentiation it is expressed at significantly higher levels in a subset of neuronal compared to non-neuronal cells. Furthermore, shRNA-mediated knockdown of MEF2A reduces the number of NSC-derived neurons compared to non-neuronal cells after differentiation. Together, these data indicate that MEF2A participates in neuronal differentiation/maturation from NSCs.


Asunto(s)
Diferenciación Celular , Factores de Transcripción MEF2/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Ratas Wistar
12.
PLoS Genet ; 13(8): e1006968, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28846707

RESUMEN

Neurotrophism, structural plasticity, learning and long-term memory in mammals critically depend on neurotrophins binding Trk receptors to activate tyrosine kinase (TyrK) signaling, but Drosophila lacks full-length Trks, raising the question of how these processes occur in the fly. Paradoxically, truncated Trk isoforms lacking the TyrK predominate in the adult human brain, but whether they have neuronal functions independently of full-length Trks is unknown. Drosophila has TyrK-less Trk-family receptors, encoded by the kekkon (kek) genes, suggesting that evolutionarily conserved functions for this receptor class may exist. Here, we asked whether Keks function together with Drosophila neurotrophins (DNTs) at the larval glutamatergic neuromuscular junction (NMJ). We tested the eleven LRR and Ig-containing (LIG) proteins encoded in the Drosophila genome for expression in the central nervous system (CNS) and potential interaction with DNTs. Kek-6 is expressed in the CNS, interacts genetically with DNTs and can bind DNT2 in signaling assays and co-immunoprecipitations. Ligand binding is promiscuous, as Kek-6 can also bind DNT1, and Kek-2 and Kek-5 can also bind DNT2. In vivo, Kek-6 is found presynaptically in motoneurons, and DNT2 is produced by the muscle to function as a retrograde factor at the NMJ. Kek-6 and DNT2 regulate NMJ growth and synaptic structure. Evidence indicates that Kek-6 does not antagonise the alternative DNT2 receptor Toll-6. Instead, Kek-6 and Toll-6 interact physically, and together regulate structural synaptic plasticity and homeostasis. Using pull-down assays, we identified and validated CaMKII and VAP33A as intracellular partners of Kek-6, and show that they regulate NMJ growth and active zone formation downstream of DNT2 and Kek-6. The synaptic functions of Kek-6 could be evolutionarily conserved. This raises the intriguing possibility that a novel mechanism of structural synaptic plasticity involving truncated Trk-family receptors independently of TyrK signaling may also operate in the human brain.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Factores de Crecimiento Nervioso/metabolismo , Plasticidad Neuronal/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas Portadoras/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Drosophila/genética , Drosophila/fisiología , Humanos , Larva/metabolismo , Neuronas Motoras/metabolismo , Factores de Crecimiento Nervioso/genética , Unión Neuromuscular , Unión Proteica , Transducción de Señal , Transmisión Sináptica
13.
J Cell Biol ; 216(5): 1421-1438, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28373203

RESUMEN

Cell number plasticity is coupled to circuitry in the nervous system, adjusting cell mass to functional requirements. In mammals, this is achieved by neurotrophin (NT) ligands, which promote cell survival via their Trk and p75NTR receptors and cell death via p75NTR and Sortilin. Drosophila NTs (DNTs) bind Toll receptors instead to promote neuronal survival, but whether they can also regulate cell death is unknown. In this study, we show that DNTs and Tolls can switch from promoting cell survival to death in the central nervous system (CNS) via a three-tier mechanism. First, DNT cleavage patterns result in alternative signaling outcomes. Second, different Tolls can preferentially promote cell survival or death. Third, distinct adaptors downstream of Tolls can drive either apoptosis or cell survival. Toll-6 promotes cell survival via MyD88-NF-κB and cell death via Wek-Sarm-JNK. The distribution of adaptors changes in space and time and may segregate to distinct neural circuits. This novel mechanism for CNS cell plasticity may operate in wider contexts.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Receptores Toll-Like/metabolismo , Animales , Recuento de Células , Muerte Celular , Supervivencia Celular , Drosophila , Plasticidad Neuronal , Transducción de Señal
14.
Int J Mol Sci ; 17(11)2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27879672

RESUMEN

The guided migration of neural cells is essential for repair in the central nervous system (CNS). Oligodendrocyte progenitor cells (OPCs) will normally migrate towards an injury site to re-sheath demyelinated axons; however the mechanisms underlying this process are not well understood. Endogenous electric fields (EFs) are known to influence cell migration in vivo, and have been utilised in this study to direct the migration of OPCs isolated from neonatal Sprague-Dawley rats. The OPCs were exposed to physiological levels of electrical stimulation, and displayed a marked electrotactic response that was dependent on ß1 integrin, one of the key subunits of integrin receptors. We also observed that F-actin, an important component of the cytoskeleton, was re-distributed towards the leading edge of the migrating cells, and that this asymmetric rearrangement was associated with ß1 integrin function.


Asunto(s)
Citoesqueleto/metabolismo , Integrina beta1/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal , Células Madre/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular , Movimiento Celular , Citoesqueleto/ultraestructura , Estimulación Eléctrica , Electricidad , Expresión Génica , Integrina beta1/genética , Células-Madre Neurales/ultraestructura , Oligodendroglía/ultraestructura , Cultivo Primario de Células , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Células Madre/ultraestructura
15.
Int J Neurosci ; 126(11): 955-62, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26824870

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons of the substantia nigra pars compacta in the brain with an unknown cause. Current pharmacological treatments for PD are only symptomatic and there is still no cure for this disease nowadays. In fact, transplantation of human fetal ventral midbrain cells into PD brains has provided a proof of concept that cell replacement therapy can be used for some PD patients, beneficial for improving their symptoms. However, the ethical and practical issues of human fetal tissue will inevitably limit its widespread clinical use. Therefore, it is essential to find alternative cell sources for the future cell transplantation for PD patients. With recent development in stem cell technology, here, we review the different types of stem cells and their main properties currently explored, which could be developed as a possible cell therapy for PD treatment.


Asunto(s)
Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Humanos , Trasplante de Células Madre/tendencias
16.
ACS Biomater Sci Eng ; 1(6): 431-439, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26240838

RESUMEN

Trauma to the central and peripheral nervous systems often lead to serious morbidity. Current surgical methods for repairing or replacing such damage have limitations. Tissue engineering offers a potential alternative. Here we show that functionalized α-helical-peptide hydrogels can be used to induce attachment, migration, proliferation and differentiation of murine embryonic neural stem cells (NSCs). Specifically, compared with undecorated gels, those functionalized with Arg-Gly-Asp-Ser (RGDS) peptides increase the proliferative activity of NSCs; promote their directional migration; induce differentiation, with increased expression of microtubule-associated protein-2, and a low expression of glial fibrillary acidic protein; and lead to the formation of larger neurospheres. Electrophysiological measurements from NSCs grown in RGDS-decorated gels indicate developmental progress toward mature neuron-like behavior. Our data indicate that these functional peptide hydrogels may go some way toward overcoming the limitations of current approaches to nerve-tissue repair.

17.
J Neuroinflammation ; 12: 109, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26025034

RESUMEN

BACKGROUND: Large-scale macrophage infiltration and reactive astrogliosis are hallmarks of early spinal cord injury (SCI) pathology. The exact nature of the macrophage response and relationship between these phenomena have not been explored in detail. Here, we have investigated these responses using a combination of in vivo SCI models, organotypic and primary cultures. METHODS: In vivo macrophage response was investigated using a contusive injury mouse model. Interactions between astrocytes and macrophages were studied in primary or organotypic cultures. Proliferation was assessed though MTT assay and nucleotide incorporation and gene expression changes through qPCR. RESULTS: Seven days following contusive SCI, a mixed M1/M2 macrophage response was seen in the injury site. Conditioned medium from primary M1, but not M2, macrophages are able to induce astrocyte proliferation in both organotypic spinal cord cultures and primary astrocytes. Soluble factors from M1 macrophages induce a reactive astrocyte gene expression pattern, whereas M2 factors inhibit expression of these genes. M2-stimulated astrocytes are also able to decrease both M1 and M2 macrophage proliferation and decrease TNFα production in M1 macrophages. CONCLUSIONS: These results suggest a strong role of M1 macrophages in inducing reactive astrogliosis and the existence of an astrocyte-mediated negative feedback system in order to dampen the immune response. These results, combined with the poor outcomes of the current immunosuppressive steroid treatments in SCI, indicate the need for more targeted therapies, taking into account the significantly different effects of M1 and M2 macrophages, in order to optimise outcome.


Asunto(s)
Astrocitos/fisiología , Comunicación Celular/fisiología , Proliferación Celular/fisiología , Gliosis/fisiopatología , Inflamación/fisiopatología , Macrófagos/fisiología , Fenotipo , Traumatismos de la Médula Espinal/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Movimiento Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Femenino , Gliosis/patología , Inmunidad Celular/fisiología , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Traumatismos de la Médula Espinal/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Exp Neurol ; 263: 113-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25265211

RESUMEN

Neural progenitor cell (NPC) replacement therapy is a promising treatment for neurodegenerative disorders including Parkinson's disease (PD). It requires a controlled directional migration and integration of NPCs, for example dopaminergic (DA) progenitor cells, into the damaged host brain tissue. There is, however, only limited understanding of how to regulate the directed migration of NPCs to the diseased or damaged brain tissues for repair and regeneration. The aims of this study are to explore the possibility of using a physiological level of electrical stimulation to regulate the directed migration of ventral midbrain NPCs (NPCs(vm)), and to investigate their potential regulation via GSK3ß and associated downstream effectors. We tested the effects of direct-current (DC) electric fields (EFs) on the migration behavior of the NPCs(vm). A DC EF induced directional cell migration toward the cathode, namely electrotaxis. Reversal of the EF polarity triggered a sharp reversal of the NPC(vm) electrotaxis. The electrotactic response was both time and EF voltage dependent. Pharmacologically inhibiting the canonical Wnt/GSK3ß pathway significantly reduced the electrotactic response of NPCs(vm), which is associated with the down-regulation of GSK3ß phosphorylation, ß-catenin activation and CLASP2 expression. This was further proved by RNA interference of GSK3ß, which also showed a significantly reduced electrotactic response in association with reduced ß-catenin activation and CLASP2 expression. In comparison, RNA interference of ß-catenin slightly reduced electrotactic response and CLASP2 expression. Both pharmacological inhibition of Wnt/GSK3ß and RNA interference of GSK3ß/ß-catenin clearly reduced the asymmetric redistribution of CLASP2 and its co-localization with α-tubulin. These results suggest that Wnt/GSK3ß signaling contributes to the electrotactic response of NPCs(vm) through the coordination of GSK3ß phosphorylation, ß-catenin activation, CLASP2 expression and asymmetric redistribution to the leading edge of the migrating cells.


Asunto(s)
Movimiento Celular/fisiología , Campos Electromagnéticos , Glucógeno Sintasa Quinasa 3/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Wnt/metabolismo , Animales , Western Blotting , Células Cultivadas , Neuronas Dopaminérgicas/metabolismo , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3 beta , Proteínas Asociadas a Microtúbulos/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transfección , beta Catenina/metabolismo
19.
Curr Protoc Stem Cell Biol ; 31: 2D.17.1-15, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25366898

RESUMEN

Oligodendrocytes are the myelinating cells of the central nervous system (CNS). The isolation of purified oligodendrocyte progenitor cells (OPCs) in large numbers has been sought after as a source of cells for repair following CNS-demyelinating diseases and injuries, such as multiple sclerosis (MS) and spinal cord injury (SCI). Methods for isolation of OPCs from rodent neonatal brains are well established and have formed the basis for research in myelin repair within the CNS for many years. However, long-term maintenance of OPCs has been a challenge owing to small cellular yields per animal and spontaneous differentiation within a short period of time. Much effort has been devoted to achieving long-term culture and maintenance of OPCs, but little progress has been made. Here, protocols are presented for preparation of highly enriched rat OPC populations and for their long-term maintenance as oligospheres using mixed-glial-conditioned medium. Functional myelinating oligodendrocytes can be achieved from such protocols, when co-cultured with primary neurons. This approach is an extension of our normal shaking method for isolating OPCs, and incorporates some adaptations from previous OPC culture methods.


Asunto(s)
Encéfalo , Separación Celular/métodos , Técnicas de Cocultivo/métodos , Vaina de Mielina/metabolismo , Oligodendroglía , Células Madre , Animales , Encéfalo/citología , Encéfalo/metabolismo , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas , Células Madre/citología , Células Madre/metabolismo
20.
PLoS One ; 8(10): e75902, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24124519

RESUMEN

Retrograde growth factors regulating synaptic plasticity at the neuromuscular junction (NMJ) in Drosophila have long been predicted but their discovery has been scarce. In vertebrates, such retrograde factors produced by the muscle include GDNF and the neurotrophins (NT: NGF, BDNF, NT3 and NT4). NT superfamily members have been identified throughout the invertebrates, but so far no functional in vivo analysis has been carried out at the NMJ in invertebrates. The NT family of proteins in Drosophila is formed of DNT1, DNT2 and Spätzle (Spz), with sequence, structural and functional conservation relative to mammalian NTs. Here, we investigate the functions of Drosophila NTs (DNTs) at the larval NMJ. All three DNTs are expressed in larval body wall muscles, targets for motor-neurons. Over-expression of DNTs in neurons, or the activated form of the Spz receptor, Toll(10b), in neurons only, rescued the semi-lethality of spz(2) and DNT1(41), DNT2(e03444) double mutants, indicating retrograde functions in neurons. In spz(2) mutants, DNT1(41), DNT2(e03444) double mutants, and upon over-expression of the DNTs, NMJ size and bouton number increased. Boutons were morphologically abnormal. Mutations in spz and DNT1,DNT2 resulted in decreased number of active zones per bouton and decreased active zone density per terminal. Alterations in DNT function induced ghost boutons and synaptic debris. Evoked junction potentials were normal in spz(2) mutants and DNT1(41), DNT2(e03444) double mutants, but frequency and amplitude of spontaneous events were reduced in spz(2) mutants suggesting defective neurotransmission. Our data indicate that DNTs are produced in muscle and are required in neurons for synaptogenesis. Most likely alterations in DNT function and synapse formation induce NMJ plasticity leading to homeostatic adjustments that increase terminal size restoring overall synaptic transmission. Data suggest that Spz functions with neuron-type specificity at the muscle 4 NMJ, and DNT1 and DNT2 function together at the muscles 6,7 NMJ.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Neuronas/metabolismo , Animales , Drosophila , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...