Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Neurochem ; 157(3): 684-694, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33251603

RESUMEN

Various lines of evidence implicate oxidative stress in the pathogenic mechanism(s) underpinning tauopathies. Consequently, antioxidant therapies have been considered in clinical practice for the treatment of tauopathies such as Alzheimer's disease (AD), but with mixed results. We and others have previously reported increased protein oxidation upon expression of both human 0N3R (hTau0N3R ) and 0N4R (hTau0N4R ) tau in vivo. Building on these studies, we demonstrate here the suppression of hTau0N3R associated phenotypes in Drosophila melanogaster after treatment with vitamin C or vitamin E. Curiously the rescue of phenotype was seen without alteration in total tau level or alteration in phosphorylation at a number of disease-associated sites. Moreover, treatment with paraquat, a pro-oxidant drug, did not exacerbate the hTau0N3R phenotypes. This result following paraquat treatment is reminiscent of our previous findings with hTau0N4R which also causes greater oxidative stress when compared to hTau0N3R but has a milder phenotype. Collectively our data imply that the role of oxidative stress in tau-mediated toxicity is not straight forward and there may be isoform-specific effects as well as contribution of other factors. This may explain the ambiguous effects of anti-oxidant treatments on clinical outcome in dementia patients.


Asunto(s)
Antioxidantes/farmacología , Vitamina E/farmacología , Proteínas tau/antagonistas & inhibidores , Proteínas tau/toxicidad , Animales , Ácido Ascórbico/farmacología , Drosophila melanogaster , Larva , Locomoción/efectos de los fármacos , Masculino , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Paraquat/toxicidad , Fosforilación/efectos de los fármacos
2.
J Biol Chem ; 294(10): 3744-3759, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30635404

RESUMEN

Extracellular vesicles (EVs) are secreted by myriad cells in culture and also by unicellular organisms, and their identification in mammalian fluids suggests that EV release also occurs at the organism level. However, although it is clearly important to better understand EVs' roles in organismal biology, EVs in solid tissues have received little attention. Here, we modified a protocol for EV isolation from primary neural cell culture to collect EVs from frozen whole murine and human neural tissues by serial centrifugation and purification on a sucrose gradient. Quantitative proteomics comparing brain-derived EVs from nontransgenic (NTg) and a transgenic amyotrophic lateral sclerosis (ALS) mouse model, superoxide dismutase 1 (SOD1)G93A, revealed that these EVs contain canonical exosomal markers and are enriched in synaptic and RNA-binding proteins. The compiled brain EV proteome contained numerous proteins implicated in ALS, and EVs from SOD1G93A mice were significantly depleted in myelin-oligodendrocyte glycoprotein compared with those from NTg animals. We observed that brain- and spinal cord-derived EVs, from NTg and SOD1G93A mice, are positive for the astrocyte marker GLAST and the synaptic marker SNAP25, whereas CD11b, a microglial marker, was largely absent. EVs from brains and spinal cords of the SOD1G93A ALS mouse model, as well as from human SOD1 familial ALS patient spinal cord, contained abundant misfolded and nonnative disulfide-cross-linked aggregated SOD1. Our results indicate that CNS-derived EVs from an ALS animal model contain pathogenic disease-causing proteins and suggest that brain astrocytes and neurons, but not microglia, are the main EV source.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Astrocitos/patología , Vesículas Extracelulares/enzimología , Neuronas/patología , Pliegue de Proteína , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/patología , Glicoproteínas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/metabolismo , Proteómica , Médula Espinal/patología , Superóxido Dismutasa-1/metabolismo
3.
Neurobiol Dis ; 105: 74-83, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28502805

RESUMEN

Tau exists as six closely related protein isoforms in the adult human brain. These are generated from alternative splicing of a single mRNA transcript and they differ in the absence or presence of two N-terminal and three or four microtubule binding domains. Typically all six isoforms have been considered functionally similar. However, their differential involvement in particular tauopathies raises the possibility that there may be isoform-specific differences in physiological function and pathological role. To explore this, we have compared the phenotypes induced by the 0N3R and 0N4R isoforms in Drosophila. Expression of the 3R isoform causes more profound axonal transport defects and locomotor impairments, culminating in a shorter lifespan than the 4R isoform. In contrast, the 4R isoform leads to greater neurodegeneration and impairments in learning and memory. Furthermore, the phosphorylation patterns of the two isoforms are distinct, as is their ability to induce oxidative stress. These differences are not consequent to different expression levels and are suggestive of bona fide physiological differences in isoform biology and pathological potential. They may therefore explain isoform-specific mechanisms of tau-toxicity and the differential susceptibility of brain regions to different tauopathies.


Asunto(s)
Modelos Animales de Enfermedad , Secuencias Repetidas en Tándem/genética , Tauopatías/genética , Tauopatías/fisiopatología , Proteínas tau/genética , Factores de Edad , Animales , Animales Modificados Genéticamente , Transporte Axonal , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Humanos , Larva/genética , Aprendizaje/fisiología , Locomoción/genética , Masculino , Memoria/fisiología , Fenotipo , Fosforilación , Isoformas de Proteínas/metabolismo , Tauopatías/mortalidad , Tauopatías/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vías Visuales/metabolismo , Proteínas tau/metabolismo
4.
Acta Neuropathol Commun ; 11(1): 200, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38111057

RESUMEN

TAR DNA-binding protein 43 (TDP-43) and Fused in Sarcoma/Translocated in Sarcoma (FUS) are ribonucleoproteins associated with pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Under physiological conditions, TDP-43 and FUS are predominantly localized in the nucleus, where they participate in transcriptional regulation, RNA splicing and metabolism. In disease, however, they are typically mislocalized to the cytoplasm where they form aggregated inclusions. A number of shared cellular pathways have been identified that contribute to TDP-43 and FUS toxicity in neurodegeneration. In the present study, we report a novel pathogenic mechanism shared by these two proteins. We found that pathological FUS co-aggregates with a ribosomal protein, the Receptor for Activated C-Kinase 1 (RACK1), in the cytoplasm of spinal cord motor neurons of ALS, as previously reported for pathological TDP-43. In HEK293T cells transiently transfected with TDP-43 or FUS mutant lacking a functional nuclear localization signal (NLS; TDP-43ΔNLS and FUSΔNLS), cytoplasmic TDP-43 and FUS induced co-aggregation with endogenous RACK1. These co-aggregates sequestered the translational machinery through interaction with the polyribosome, accompanied by a significant reduction of global protein translation. RACK1 knockdown decreased cytoplasmic aggregation of TDP-43ΔNLS or FUSΔNLS and alleviated associated global translational suppression. Surprisingly, RACK1 knockdown also led to partial nuclear localization of TDP-43ΔNLS and FUSΔNLS in some transfected cells, despite the absence of NLS. In vivo, RACK1 knockdown alleviated retinal neuronal degeneration in transgenic Drosophila melanogaster expressing hTDP-43WT or hTDP-43Q331K and improved motor function of hTDP-43WT flies, with no observed adverse effects on neuronal health in control knockdown flies. In conclusion, our results revealed a novel shared mechanism of pathogenesis for misfolded aggregates of TDP-43 and FUS mediated by interference with protein translation in a RACK1-dependent manner. We provide proof-of-concept evidence for targeting RACK1 as a potential therapeutic approach for TDP-43 or FUS proteinopathy associated with ALS and FTLD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Sarcoma , Animales , Humanos , Esclerosis Amiotrófica Lateral/patología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Neuronas Motoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/patología , Degeneración Lobar Frontotemporal/patología , Biosíntesis de Proteínas , Sarcoma/metabolismo , Sarcoma/patología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética
5.
Biochem Biophys Res Commun ; 422(3): 351-7, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22580275

RESUMEN

NELL-1 is a potent osteoinductive molecule that enhances bone formation in multiple animal models through currently unidentified pathways. In the present manuscript, we hypothesized that NELL-1 may regulate osteogenic differentiation accompanied by alteration of inorganic phosphate (Pi) entry into the osteoblast via sodium dependent phosphate (NaPi) transporters. To determine this, MC3T3-E1 pre-osteoblasts were cultured in the presence of recombinant human (rh)NELL-1 or rhBMP-2. Analysis was performed for intracellular Pi levels through malachite green staining, Pit-1 and Pit-2 expression, and forced upregulation of Pit-1 and Pit-2. Results showed rhNELL-1 to increase MC3T3-E1 matrix mineralization and Pi influx associated with activation of both Pit-1 and Pit-2 channels, with significantly increased Pit-2 production. In contrast, Pi transport elicited by rhBMP-2 showed to be associated with increased Pit-1 production only. Next, neutralizing antibodies against Pit-1 and Pit-2 completely abrogated the Pi influx effect of rhNELL-1, suggesting rhNELL-1 is dependent on both transporters. These results identify one potential mechanism of action for rhNELL-1 induced osteogenesis and highlight a fundamental difference between NELL-1 and BMP-2 signaling.


Asunto(s)
Calcificación Fisiológica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/fisiología , Osteogénesis/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Proteínas de Unión al Calcio , Línea Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Transducción de Señal
6.
Biochem Soc Trans ; 40(4): 693-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22817718

RESUMEN

Insoluble aggregates of the microtubule-associated protein tau characterize a number of neurodegenerative diseases collectively termed tauopathies. These aggregates comprise abnormally hyperphosphorylated and misfolded tau proteins. Research in this field has traditionally focused on understanding how hyperphosphorylated and aggregated tau mediates dysfunction and toxicity in tauopathies. Recent findings from both Drosophila and rodent models of tauopathy suggest that large insoluble aggregates such as tau filaments and tangles may not be the key toxic species in these diseases. Thus some investigators have shifted their focus to study pre-filament tau species such as tau oligomers and hyperphosphorylated tau monomers. Interestingly, tau oligomers can exist in a variety of states including hyperphosphorylated and unphosphorylated forms, which can be both soluble and insoluble. It remains to be determined which of these oligomeric states of tau are causally involved in neurodegeneration and which signal the beginning of the formation of inert/protective filaments. It will be important to better understand this so that tau-based therapeutic interventions can target the most toxic tau species.


Asunto(s)
Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Humanos , Fosforilación , Tauopatías/metabolismo
7.
J Neurosci ; 29(35): 10928-38, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19726651

RESUMEN

Evidence suggests that NMDA-type glutamate receptors contribute to degeneration of striatal medium-sized spiny neurons (MSNs) in Huntington's disease (HD). Previously, we demonstrated that NMDA receptor (NMDAR)-mediated current and/or toxicity is increased in MSNs from the yeast artificial chromosome (YAC) transgenic mouse model expressing polyglutamine (polyQ)-expanded (mutant) full-length human huntingtin (htt). Others have shown that membrane-associated guanylate kinases (MAGUKs), such as PSD-95 and SAP102, modulate NMDAR surface expression and excitotoxicity in hippocampal and cortical neurons and that htt interacts with PSD-95. Here, we tested the hypothesis that an altered association between MAGUKs and NMDARs in mutant huntingtin-expressing cells contributes to increased susceptibility to excitotoxicity. We show that htt coimmunoprecipitated with SAP102 in HEK293T cells and striatal tissue from wild-type and YAC transgenic mice; however, the association of SAP102 with htt or the NMDAR NR2B subunit was unaffected by htt polyQ length, whereas association of PSD-95 with NR2B in striatal tissue was enhanced by increased htt polyQ length. Treatment of cultured MSNs with Tat-NR2B9c peptide blocked binding of NR2B with SAP102 and PSD-95 and reduced NMDAR surface expression by 20% in both YAC transgenic and wild-type MSNs, and also restored susceptibility to NMDAR excitoxicity in YAC HD MSNs to levels observed in wild-type MSNs; a similar effect on excitotoxicity was observed after knockdown of PSD-95 by small interfering RNA. Unlike previous findings in cortical and hippocampal neurons, rescue of NMDA toxicity by Tat-NR2B9c occurred independently of any effect on neuronal nitric oxide synthase activity. Our results elucidate further the mechanisms underlying enhanced excitotoxicity in HD.


Asunto(s)
Cromosomas Artificiales de Levadura/metabolismo , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/metabolismo , Enfermedad de Huntington/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Línea Celular , Células Cultivadas , Cromosomas Artificiales de Levadura/genética , Homólogo 4 de la Proteína Discs Large , Predisposición Genética a la Enfermedad , Guanilato-Quinasas , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Receptores de N-Metil-D-Aspartato/genética
8.
Biochem Soc Trans ; 38(4): 988-92, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20658990

RESUMEN

AD (Alzheimer's disease) is a neurodegenerative disorder characterized by the abnormal hyperphosphorylation and aggregation of the microtubule-associated protein tau and the misfolding and deposition of Abeta peptide. The mechanisms by which tau and Abeta become abnormal is not clearly understood, neither is it known what role either protein plays in the neurodegenerative process underlying AD. We have modelled aspects of AD in Drosophila melanogaster to shed light on these processes and to further our understanding of the relationship between tau and amyloid in this disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Drosophila melanogaster , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/fisiología , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Humanos , Modelos Biológicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Péptido Hidrolasas/metabolismo , Fosforilación , Transmisión Sináptica/fisiología , Proteínas tau/metabolismo , Proteínas tau/fisiología
9.
Biochem Soc Trans ; 38(2): 564-70, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20298222

RESUMEN

Axonal microtubules are essential for transport of materials to the synapse. Compromised microtubules and synaptic loss have been demonstrated in AD (Alzheimer's disease), which is believed to contribute to cognitive dysfunction before neuronal death in the early stages of the disease. The mechanism by which hyperphosphorylated tau, the building block of neurofibrillary tangles, one of the pathological hallmarks of AD, disrupts neuronal and synaptic function is unclear. There is a theory that hyperphosphorylated tau does not bind effectively to microtubules and is no longer able to function in stabilizing them, thus axonal transport can no longer proceed efficiently. This leads to synaptic dysfunction. We have tested this theory in a Drosophila model of tauopathies in which we expressed human tau (h-tau). Using this model, we have tested all aspects of this hypothesis and have demonstrated that axonal transport does become compromised in the presence of hyperphosphorylated h-tau and this leads to synaptic and behavioural defects. We are currently investigating the mechanism by which hyperphosphorylated h-tau mediates this effect and are preliminary data indicate that this entails phospho-tau-mediated effects that are predicted by the tau-microtubule hypothesis, as well as novel effects. These deleterious effects of h-tau occur in the absence of tau filaments and before neuronal death. This sequence of pathogenic events may constitute the mechanism by which abnormal tau disrupts neuronal and synaptic function and contributes to cognitive impairment before neuronal death in the early stages of tauopathies such as AD.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Neuronas/efectos de los fármacos , Tauopatías/patología , Proteínas tau/farmacología , Animales , Drosophila/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Modelos Biológicos , Neuronas/fisiología , Fosforilación/fisiología , Solubilidad , Tauopatías/metabolismo , Tauopatías/fisiopatología , Regulación hacia Arriba/fisiología , Proteínas tau/metabolismo
10.
Acta Neuropathol ; 120(5): 593-604, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20617325

RESUMEN

It has been hypothesised that tau protein, when hyper-phosphorylated as in Alzheimer's disease (AD), does not bind effectively to microtubules and is no longer able to stabilise them; thus microtubules break down, and axonal transport can no longer proceed efficiently in affected brain regions in AD and related tauopathies (tau-microtubule hypothesis). We have used Drosophila models of tauopathy to test all components of this hypothesis in vivo. We have previously shown that upon expression of human 0N3R tau in Drosophila motor neurons it becomes highly phosphorylated, resulting in disruptions to both axonal transport and synaptic function which culminate in behavioural phenotypes. We now show that the mechanism by which the human tau mediates these effects is twofold: first, as predicted by the tau-microtubule hypothesis, the highly phosphorylated tau exhibits significantly reduced binding to microtubules; and second, it participates in a pathogenic interaction with the endogenous normal Drosophila tau and sequesters it away from microtubules. This causes disruption of the microtubular cytoskeleton as evidenced by a reduction in the numbers of intact correctly-aligned microtubules and the appearance of microtubules that are not correctly oriented within the axon. These deleterious effects of human tau are phosphorylation dependent because treatment with LiCl to suppress tau phosphorylation increases microtubule binding of both human and Drosophila tau and restores cytoskeletal integrity. Notably, all these phospho-tau-mediated phenotypes occur in the absence of tau filament/neurofibrillary tangle formation or neuronal death, and may thus constitute the mechanism by which hyper-phosphorylated tau disrupts neuronal function and contributes to cognitive impairment prior to neuronal death in the early stages of tauopathies.


Asunto(s)
Microtúbulos/metabolismo , Microtúbulos/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteínas tau/metabolismo , Animales , Western Blotting , Drosophila , Humanos , Inmunohistoquímica , Inmunoprecipitación , Microscopía Electrónica de Transmisión , Fosforilación , Tauopatías/metabolismo , Tauopatías/patología
11.
J Oral Maxillofac Surg ; 68(2): 300-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20116699

RESUMEN

PURPOSE: Most craniofacial birth defects contain skeletal components that require bone grafting. Although many growth factors have shown potential for use in bone regeneration, bone morphogenetic proteins (BMPs) are the most osteoinductive. However, supraphysiologic doses, high cost, and potential adverse effects stimulate clinicians and researchers to identify complementary molecules that allow a reduction in dose of BMP-2. Because NELL1 plays a key role as a regulator of craniofacial skeletal morphogenesis, especially in committed chondrogenic and osteogenic differentiation, and a previous synergistic mechanism has been identified, NELL1 is an ideal molecule for combination with BMP-2 in calvarial defect regeneration. We investigated the effect of NELL1 and BMP-2 on bone regeneration in vivo. MATERIALS AND METHODS: BMP-2 doses of 589 and 1,178 ng were grafted into 5-mm critical-sized rat calvarial defects, as compared with 589 ng of NELL1 plus 589 ng of BMP-2 and 1,178 ng of NELL1 plus 1,178 ng of BMP-2, and bone regeneration was analyzed. RESULTS: Live micro-computed tomography data showed increased bone formation throughout 4 to 8 weeks in all groups but a significant improvement when the lower doses of each molecule were combined. High-resolution micro-computed tomography and histology showed more mature and complete defect healing when the combination of NELL1 plus BMP-2 was compared with BMP-2 alone at lower doses. CONCLUSION: The observed potential synergy has significant value in the future treatment of patients with craniofacial defects requiring extensive bone grafting that would normally entail extraoral autogenous bone grafts or doses of BMP-2 in milligrams.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Regeneración Ósea/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta/farmacología , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/administración & dosificación , Relación Dosis-Respuesta a Droga , Portadores de Fármacos , Sinergismo Farmacológico , Humanos , Ácido Láctico , Masculino , Proteínas del Tejido Nervioso/administración & dosificación , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Cráneo/cirugía , Factor de Crecimiento Transformador beta/administración & dosificación , Microtomografía por Rayos X
12.
Nat Neurosci ; 9(6): 824-31, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699508

RESUMEN

Post-translational modification by the lipid palmitate is crucial for the correct targeting and function of many proteins. Here we show that huntingtin (htt) is normally palmitoylated at cysteine 214, which is essential for its trafficking and function. The palmitoylation and distribution of htt are regulated by the palmitoyl transferase huntingtin interacting protein 14 (HIP14). Expansion of the polyglutamine tract of htt, which causes Huntington disease, results in reduced interaction between mutant htt and HIP14 and consequently in a marked reduction in palmitoylation. Mutation of the palmitoylation site of htt, making it palmitoylation resistant, accelerates inclusion formation and increases neuronal toxicity. Downregulation of HIP14 in mouse neurons expressing wild-type and mutant htt increases inclusion formation, whereas overexpression of HIP14 substantially reduces inclusions. These results suggest that the expansion of the polyglutamine tract in htt results in decreased palmitoylation, which contributes to the formation of inclusion bodies and enhanced neuronal toxicity.


Asunto(s)
Proteínas Portadoras/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Ácido Palmítico/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos/fisiología , Animales , Animales Recién Nacidos , Células COS , Proteínas Portadoras/genética , Células Cultivadas , Corteza Cerebral/citología , Chlorocebus aethiops , Cisteína/metabolismo , Regulación hacia Abajo/genética , Humanos , Proteína Huntingtina , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Transporte de Proteínas/fisiología , Ratas , Expansión de Repetición de Trinucleótido/genética
13.
J Neurosci ; 28(48): 12725-35, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19036965

RESUMEN

Huntington disease (HD), caused by CAG expansion in the ubiquitously expressed huntingtin gene, is characterized by early dysfunction and death of striatal medium-sized spiny neurons (MSNs). Previous work has shown MSN-specific alterations in NMDA receptor (NMDAR) expression and cell death signaling. Furthermore, studies in HD human brain tissue and a knock-in mouse model demonstrate increases in calpain activity, which can be stimulated by NMDARs and contribute to excitotoxicity. Here, we report increased calpain activity in MSNs from the yeast artificial chromosome (YAC) transgenic mouse model of HD, expressing human full-length huntingtin with 128 polyglutamine repeats (YAC128), compared with wild type. Moreover, the calpain-cleaved product of NMDAR subunit NR2B is increased early, and NR2B expression levels are reduced, in YAC128 striatum. Although steady-state NMDAR surface expression is similar in wild-type and YAC128 MSNs, the rate of loss of NR2B-containing surface receptors is enhanced in YAC128 MSNs, suggesting that NMDAR forward trafficking to the surface is also faster, as previously reported for YAC72 MSNs. Calpain inhibitor-1 treatment normalized the loss rate of surface NMDARs in YAC128 MSNs to that of wild type, and significantly increased surface NMDAR expression in YAC128, but not in wild type or YAC72. With acute NMDAR overstimulation, the increase in calpain activity correlated with polyglutamine length, and calpain inhibitor treatment reduced NMDA-induced apoptosis in YAC72 and YAC128 MSNs to wild-type levels. Thus, the cumulative effect of increasing huntingtin polyglutamine length is to enhance MSN sensitivity to excitotoxicity at least in part by calpain-mediated cell death signaling.


Asunto(s)
Calpaína/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Glicoproteínas/farmacología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/genética , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Expansión de Repetición de Trinucleótido/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Levaduras/genética , Levaduras/metabolismo
14.
Biol Proced Online ; 11: 161-9, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19495917

RESUMEN

The dissociation of adherent mesenchymal stem cell (MSC) monolayers with trypsin and enzyme-free dissociation buffer was compared. A significantly lower proportion of viable cells were obtained with enzyme-free dissociation buffers compared to trypsin. Subsequently, the dissociated cells were re-seeded on new cell culture dishes and were subjected to the MTT assay 24 h later. The proportion of viable cells that reattached was significantly lower for cells obtained by dissociation with enzyme-free dissociation buffer compared to trypsin. Frozen-thawed MSC displayed a similar trend, yielding consistently higher cell viability and reattachment rates when dissociated with trypsin compared to enzyme-free dissociation buffer. It was also demonstrated that exposure of trypsin-dissociated MSC to enzyme-free dissociation buffer for 1 h had no significant detrimental effect on cell viability.

15.
ACS Chem Neurosci ; 9(7): 1591-1606, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29614860

RESUMEN

Oligomers of amyloid-ß (AßO) are deemed key in synaptotoxicity and amyloid seeding of Alzheimer's disease (AD). However, the heterogeneous and dynamic nature of AßO and inadequate markers for AßO subtypes have stymied effective AßO identification and therapeutic targeting in vivo. We identified an AßO-subclass epitope defined by differential solvent orientation of the lysine 28 side chain in a constrained loop of serine-asparagine-lysine (cSNK), rarely displayed in molecular dynamics simulations of monomer and fibril ensembles. A mouse monoclonal antibody targeting AßOcSNK recognizes ∼50-60 kDa SDS-resistant soluble Aß assemblages in AD brain and prolongs the lag phase of Aß aggregation in vitro. Acute peripheral infusion of a murine IgG1 anti-AßOcSNK in two AD mouse models reduced soluble brain Aß aggregates by 20-30%. Chronic cSNK peptide immunization of APP/PS1 mice engendered an anti-AßOcSNK IgG1 response without epitope spreading to Aß monomers or fibrils and was accompanied by preservation of global PSD95 expression and improved cued fear memory. Our data indicate that the oligomer subtype AßOcSNK participates in synaptotoxicity and propagation of Aß aggregation in vitro and in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/inmunología , Epítopos , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Animales , Encéfalo/inmunología , Encéfalo/patología , Química Encefálica , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Memoria/fisiología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación de Dinámica Molecular , Placa Amiloide/química , Placa Amiloide/inmunología , Placa Amiloide/patología , Agregación Patológica de Proteínas , Conformación Proteica , Multimerización de Proteína
16.
J Bone Miner Res ; 22(6): 918-30, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17352654

RESUMEN

UNLABELLED: Osteogenesis is synergistically enhanced by the combined effect of complimentary factors. This study showed that Nell-1 and BMP-2 synergistically enhanced osteogenic differentiation of myoblasts and phosphorylated the JNK MAPK pathway. The findings are important because of the osteochondral specificity of Nell-1 signaling and the potential therapeutic effects of coordinated BMP-2 and Nell-1 delivery. INTRODUCTION: BMPs play an important role in the migration and proliferation of mesenchymal cells and have a unique ability to alter the differentiation of mesenchymal cells toward chondrogenic and osteogenic lineages. Signaling upstream of Cbfa1/Runx2, BMPs effects are not limited to cells of the osteoblast lineage. Thus, additional osteoblast-specific factors that could synergize with BMP-2 would be advantageous for bone regeneration procedures. NELL-1 (NEL-like molecule-1; NEL [a protein strongly expressed in neural tissue encoding epidermal growth factor like domain]) is a novel growth factor believed to preferentially target cells committed to the osteochondral lineage. MATERIALS AND METHODS: C2C12 myoblasts were transduced with AdLacZ, AdNell-1, AdBMP-2, or AdNell-1+AdBMP-2 overexpression viruses. Effects were studied by cell morphology, alkaline phosphatase activity, osteopontin production, and MAPK signaling. Additionally, in a nude mouse model, viruses were injected into leg muscles, and new bone formation was examined after 2 and 8 wk. RESULTS: C2C12 myoblasts co-transduced with AdNell-1+AdBMP-2 showed a synergistic effect on osteogenic differentiation as detected by alkaline phosphatase activity and osteopontin production. Nell-1 stimulation on AdNell-1 + AdBMP-2 preconditioned C2C12 cells revealed significant activation of the non-BMP-2 associated c-Jun N-terminal kinase (JNK) MAPK signaling pathway, but not the p38 or extracellular signal-regulated kinase (ERK1/2) MAPK pathways. Importantly Nell-1 alone did not induce osteogenic differentiation of myoblasts. In a nude mouse model, injection of AdNell-1 alone stimulated no bone formation within muscle; however, injection of AdNell-1+AdBMP-2 stimulated a synergistic increase in bone formation compared with AdBMP-2 alone. CONCLUSIONS: These findings are important because of the confirmed osteochondral specificity of Nell-1 signaling and the potential therapeutic effects of enhanced BMP-2 action with coordinated Nell-1 delivery.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/fisiología , Mioblastos/metabolismo , Proteínas del Tejido Nervioso/fisiología , Factor de Crecimiento Transformador beta/fisiología , Adenoviridae/genética , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas del Grupo de Alta Movilidad/análisis , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Desnudos , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Mioblastos/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Osteopontina/metabolismo , Ratas , Factor de Transcripción SOX9 , Transducción de Señal/fisiología , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo , Transfección , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Tissue Eng ; 13(3): 501-12, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17319794

RESUMEN

Numerous growth factors, peptides, and small molecules are being developed for bone tissue engineering. The optimal dosing, stability, and bioactivity of these biological molecules are likely influenced by the carrier biomaterial. Efficient evaluation of various formulations will require objective evaluation of in vitro culture systems and in vivo regeneration models. The objective of this paper is to examine the utility of microcomputed tomography (microCT) over conventional techniques in the evaluation of the bone morphogenetic protein-2 (BMP-2) dose response effect in a three-dimensional (3D) in vitro culture system and in an established calvarial defect model. Cultured MC3T3-E1 osteoblasts displayed increased cellular density, extracellular matrix (ECM) production, and mineralization on 3D poly(lactic-co-glycolic acid) (PLGA) scaffolds in a BMP-2 dose dependent manner. MicroCT revealed differences in shape and spatial organization of mineralized areas, which would not have been possible through conventional alizarin red staining alone. Additionally, BMP-2 (doses of 30 to 240 ng/mm(3)) was grafted into 5 mm critical sized rat calvarial defects, where increased bone regeneration was observed in a dose dependent manner, with higher doses of BMP-2 inducing greater bone area, volume, and density. The data revealed the utility of microCT analysis as a beneficial addition to existing techniques for objective evaluation of bone tissue engineering and regeneration.


Asunto(s)
Proteínas Morfogenéticas Óseas/administración & dosificación , Calcificación Fisiológica/fisiología , Cráneo/lesiones , Cráneo/metabolismo , Tomografía Computarizada por Rayos X , Factor de Crecimiento Transformador beta/administración & dosificación , Células 3T3 , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/fisiología , Regeneración Ósea/fisiología , Sustitutos de Huesos , Células Cultivadas , Ratones , Ratas , Ratas Sprague-Dawley , Tomografía Computarizada por Rayos X/métodos , Factor de Crecimiento Transformador beta/fisiología
18.
J Orthop Res ; 25(11): 1488-97, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17568450

RESUMEN

Oxysterols, naturally occurring cholesterol oxidation products, can induce osteoblast differentiation. Here, we investigated short-term 22(S)-hydroxycholesterol + 20(S)-hydroxycholesterol (SS) exposure on osteoblastic differentiation of marrow stromal cells. We further explored oxysterol ability to promote bone healing in vivo. Osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity, osteocalcin (OCN) mRNA expression, mineralization, and Runx2 DNA binding activity. To explore the effects of osteogenic oxysterols in vivo, we utilized the critical-sized rat calvarial defect model. Poly(lactic-co-glycolic acid) (PLGA) scaffolds alone or coated with 140 ng (low dose) or 1400 ng (high dose) oxysterol cocktail were implanted into the defects. Rats were sacrificed at 6 weeks and examined by three-dimensional (3D) microcomputed tomography (microCT). Bone volume (BV), total volume (TV), and BV/TV ratio were measured. Culture exposure to SS for 10 min significantly increased ALP activity after 4 days, while 2 h exposure significantly increased mineralization after 14 days. Four-hour SS treatment increased OCN mRNA measured after 8 days and nuclear protein binding to an OSE2 site measured after 4 days. The calvarial defects showed slight bone healing in the control group. However, scaffolds adsorbed with low or high-dose oxysterol cocktail significantly enhanced bone formation. Histologic examination confirmed bone formation in the defect sites grafted with oxysterol-adsorbed scaffolds, compared to mostly fibrous tissue in control sites. Our results suggest that brief exposure to osteogenic oxysterols triggered events leading to osteoblastic cell differentiation and function in vitro and bone formation in vivo. These results identify oxysterols as potential agents in local and systemic enhancement of bone formation.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hidroxicolesteroles/farmacología , Osteoblastos/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea , Regeneración Ósea/fisiología , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/fisiología , Línea Celular , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Ácido Láctico/administración & dosificación , Masculino , Ratones , Osteoblastos/citología , Osteocalcina/genética , Osteocalcina/metabolismo , Ácido Poliglicólico/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/administración & dosificación , Ratas , Ratas Sprague-Dawley , Cráneo/efectos de los fármacos , Cráneo/lesiones , Cráneo/patología , Células del Estroma
19.
Nat Biotechnol ; 22(5): 560-7, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15077117

RESUMEN

In adults and children over two years of age, large cranial defects do not reossify successfully, posing a substantial biomedical burden. The osteogenic potential of bone marrow stromal (BMS) cells has been documented. This study investigates the in vivo osteogenic capability of adipose-derived adult stromal (ADAS) cells, BMS cells, calvarial-derived osteoblasts and dura mater cells to heal critical-size mouse calvarial defects. Implanted, apatite-coated, PLGA scaffolds seeded with ADAS or BMS cells produced significant intramembranous bone formation by 2 weeks and areas of complete bony bridging by 12 weeks as shown by X-ray analysis, histology and live micromolecular imaging. The contribution of implanted cells to new bone formation was 84-99% by chromosomal detection. These data show that ADAS cells heal critical-size skeletal defects without genetic manipulation or the addition of exogenous growth factors.


Asunto(s)
Tejido Adiposo/citología , Cráneo/citología , Células del Estroma/citología , Animales , Células de la Médula Ósea/citología , Ácido Láctico , Ratones , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros
20.
Nat Neurosci ; 20(4): 540-549, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28192395

RESUMEN

Drugs of abuse alter synaptic connections in the reward circuitry of the brain, which leads to long-lasting behavioral changes that underlie addiction. Here we show that cadherin adhesion molecules play a critical role in mediating synaptic plasticity and behavioral changes driven by cocaine. We demonstrate that cadherin is essential for long-term potentiation in the ventral tegmental area and is recruited to the synaptic membranes of excitatory synapses onto dopaminergic neurons following cocaine-mediated behavioral conditioning. Furthermore, we show that stabilization of cadherin at the membrane of these synapses blocks cocaine-induced synaptic plasticity, leading to a reduction in conditioned place preference induced by cocaine. Our findings identify cadherins and associated molecules as targets of interest for understanding pathological plasticity associated with addiction.


Asunto(s)
Cadherinas/fisiología , Cocaína/farmacología , Condicionamiento Psicológico/fisiología , Plasticidad Neuronal/fisiología , Área Tegmental Ventral/fisiología , Animales , Cadherinas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Receptores AMPA/metabolismo , Sinapsis/fisiología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA