Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 20(7): 4434-4460, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38779814

RESUMEN

INTRODUCTION: Tropomyosin related kinase B (TrkB) and C (TrkC) receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid beta (Aß) toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. METHODS: PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APPL/S) and wild-type controls. Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA sequencing. RESULTS: In APPL/S mice, BD10-2 treatment improved memory and LTP deficits. This was accompanied by normalized phosphorylation of protein kinase B (Akt), calcium-calmodulin-dependent kinase II (CaMKII), and AMPA-type glutamate receptors containing the subunit GluA1; enhanced activity-dependent recruitment of synaptic proteins; and increased excitatory synapse number. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. DISCUSSION: BD10-2 prevented APPL/S/Aß-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response. HIGHLIGHTS: Small molecule modulation of tropomyosin related kinase B (TrkB) and C (TrkC) restores long-term potentiation (LTP) and behavior in an Alzheimer's disease (AD) model. Modulation of TrkB and TrkC regulates synaptic activity-dependent transcription. TrkB and TrkC receptors are candidate targets for translational therapeutics. Electrophysiology combined with transcriptomics elucidates synaptic restoration. LTP identifies neuron and microglia AD-relevant human-mouse co-expression modules.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Receptor trkB , Sinapsis , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Ratones , Receptor trkB/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Sinapsis/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Receptor trkC/metabolismo , Receptor trkC/genética , Transcriptoma/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Masculino
2.
Neurobiol Dis ; 162: 105563, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838668

RESUMEN

Degeneration of basal forebrain cholinergic neurons (BFCNs) in the nucleus basalis of Meynert (NBM) and vertical diagonal band (VDB) along with their connections is a key pathological event leading to memory impairment in Alzheimer's disease (AD). Aberrant neurotrophin signaling via Trks and the p75 neurotrophin receptor (p75NTR) contributes importantly to BFCN dystrophy. While NGF/TrkA signaling has received the most attention in this regard, TrkB and TrkC signaling also provide trophic support to BFCNs and these receptors may be well located to preserve BFCN connectivity. We previously identified a small molecule TrkB/TrkC ligand, LM22B-10, that promotes cell survival and neurite outgrowth in vitro and activates TrkB/TrkC signaling in the hippocampus of aged mice when given intranasally, but shows poor oral bioavailability. An LM22B-10 derivative, PTX-BD10-2, with improved oral bioavailability has been developed and this study examined its effects on BFCN atrophy in the hAPPLond/Swe (APPL/S) AD mouse model. Oral delivery of PTX-BD10-2 was started after appreciable amyloid and cholinergic pathology was present to parallel the clinical context, as most AD patients start treatment at advanced pathological stages. PTX-BD10-2 restored cholinergic neurite integrity in the NBM and VDB, and reduced NBM neuronal atrophy in symptomatic APPL/S mice. Dystrophy of cholinergic neurites in BF target regions, including the cortex, hippocampus, and amygdala, was also reduced with treatment. Finally, PTX-BD10-2 reduced NBM tau pathology and improved the survival of cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) after amyloid-ß exposure. These data provide evidence that targeting TrkB and TrkC signaling with PTX-BD10-2 may be an effective disease-modifying strategy for combating cholinergic dysfunction in AD. The potential for clinical translation is further supported by the compound's reduction of AD-related degenerative processes that have progressed beyond early stages and its neuroprotective effects in human iPSC-derived cholinergic neurons.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Enfermedad de Alzheimer/patología , Animales , Atrofia/patología , Neuronas Colinérgicas/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Ratones , Factores de Crecimiento Nervioso , Receptor trkC , Receptores de Factor de Crecimiento Nervioso
3.
Hum Mol Genet ; 27(16): 2893-2912, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29860333

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder that has no cure. HD therapeutic development would benefit from a non-invasive translatable biomarker to track disease progression and treatment response. A potential biomarker is using positron emission tomography (PET) imaging with a translocator protein 18 kDa (TSPO) radiotracer to detect microglial activation, a key contributor to HD pathogenesis. The ability of TSPO-PET to identify microglial activation in HD mouse models, essential for a translatable biomarker, or therapeutic efficacy in HD patients or mice is unknown. Thus, this study assessed the feasibility of utilizing PET imaging with the TSPO tracer, [18F]PBR06, to detect activated microglia in two HD mouse models and to monitor response to treatment with LM11A-31, a p75NTR ligand known to reduce neuroinflammation in HD mice. [18F]PBR06-PET detected microglial activation in striatum, cortex and hippocampus of vehicle-treated R6/2 mice at a late disease stage and, notably, also in early and mid-stage symptomatic BACHD mice. After oral administration of LM11A-31 to R6/2 and BACHD mice, [18F]PBR06-PET discerned the reductive effects of LM11A-31 on neuroinflammation in both HD mouse models. [18F]PBR06-PET signal had a spatial distribution similar to ex vivo brain autoradiography and correlated with microglial activation markers: increased IBA-1 and TSPO immunostaining/blotting and striatal levels of cytokines IL-6 and TNFα. These results suggest that [18F]PBR06-PET is a useful surrogate marker of therapeutic efficacy in HD mice with high potential as a translatable biomarker for preclinical and clinical HD trials.


Asunto(s)
Corteza Cerebral/diagnóstico por imagen , Enfermedad de Huntington/diagnóstico por imagen , Receptores de GABA/administración & dosificación , Receptores de Factor de Crecimiento Nervioso/genética , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fluorodesoxiglucosa F18/administración & dosificación , Fluorodesoxiglucosa F18/química , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Isoleucina/administración & dosificación , Isoleucina/análogos & derivados , Masculino , Ratones , Microglía/efectos de los fármacos , Morfolinas/administración & dosificación , Tomografía de Emisión de Positrones , Unión Proteica , Receptores de GABA/química , Receptores de GABA/genética
4.
Hum Mol Genet ; 25(22): 4920-4938, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28171570

RESUMEN

Decreases in the ratio of neurotrophic versus neurodegenerative signalling play a critical role in Huntington's disease (HD) pathogenesis and recent evidence suggests that the p75 neurotrophin receptor (NTR) contributes significantly to disease progression. p75NTR signalling intermediates substantially overlap with those promoting neuronal survival and synapse integrity and with those affected by the mutant huntingtin (muHtt) protein. MuHtt increases p75NTR-associated deleterious signalling and decreases survival signalling suggesting that p75NTR could be a valuable therapeutic target. This hypothesis was investigated by examining the effects of an orally bioavailable, small molecule p75NTR ligand, LM11A-31, on HD-related neuropathology in HD mouse models (R6/2, BACHD). LM11A-31 restored striatal AKT and other pro-survival signalling while inhibiting c-Jun kinase (JNK) and other degenerative signalling. Normalizing p75NTR signalling with LM11A-31 was accompanied by reduced Htt aggregates and striatal cholinergic interneuron degeneration as well as extended survival in R6/2 mice. The p75NTR ligand also decreased inflammation, increased striatal and hippocampal dendritic spine density, and improved motor performance and cognition in R6/2 and BACHD mice. These results support small molecule modulation of p75NTR as an effective HD therapeutic strategy. LM11A-31 has successfully completed Phase I safety and pharmacokinetic clinical trials and is therefore a viable candidate for clinical studies in HD.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Isoleucina/análogos & derivados , Morfolinas/farmacología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Isoleucina/farmacología , Ligandos , Masculino , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Fenotipo , Unión Proteica , Distribución Aleatoria , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos
5.
J Neurosci ; 33(48): 18712-27, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24285878

RESUMEN

Loss of neurotrophic support in the striatum caused by reduced brain-derived neurotrophic factor (BDNF) levels plays a critical role in Huntington's disease (HD) pathogenesis. BDNF acts via TrkB and p75 neurotrophin receptors (NTR), and restoring its signaling is a prime target for HD therapeutics. Here we sought to determine whether a small molecule ligand, LM22A-4, specific for TrkB and without effects on p75(NTR), could alleviate HD-related pathology in R6/2 and BACHD mouse models of HD. LM22A-4 was administered to R6/2 mice once daily (5-6 d/week) from 4 to 11 weeks of age via intraperitoneal and intranasal routes simultaneously to maximize brain levels. The ligand reached levels in the R6/2 forebrain greater than the maximal neuroprotective dose in vitro and corrected deficits in activation of striatal TrkB and its key signaling intermediates AKT, PLCγ, and CREB. Ligand-induced TrkB activation was associated with a reduction in HD pathologies in the striatum including decreased DARPP-32 levels, neurite degeneration of parvalbumin-containing interneurons, inflammation, and intranuclear huntingtin aggregates. Aggregates were also reduced in the cortex. Notably, LM22A-4 prevented deficits in dendritic spine density of medium spiny neurons. Moreover, R6/2 mice given LM22A-4 demonstrated improved downward climbing and grip strength compared with those given vehicle, though these groups had comparable rotarod performances and survival times. In BACHD mice, long-term LM22A-4 treatment (6 months) produced similar ameliorative effects. These results support the hypothesis that targeted activation of TrkB inhibits HD-related degenerative mechanisms, including spine loss, and may provide a disease mechanism-directed therapy for HD and other neurodegenerative conditions.


Asunto(s)
Benzamidas/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Trastornos del Movimiento/tratamiento farmacológico , Receptor trkB/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Benzamidas/farmacocinética , Western Blotting , Peso Corporal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/fisiología , Espinas Dendríticas/fisiología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Inmunohistoquímica , Ligandos , Masculino , Ratones , Ratones Mutantes Neurológicos , Ratones Transgénicos , Trastornos del Movimiento/patología , Trastornos del Movimiento/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Equilibrio Postural/efectos de los fármacos , ARN/biosíntesis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor trkB/fisiología , Transducción de Señal/efectos de los fármacos , Sobrevida
6.
J Huntingtons Dis ; 12(3): 215-239, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37638447

RESUMEN

BACKGROUND: Loss of neurotrophic support in the striatum, particularly reduced brain-derived neurotrophic factor (BDNF) levels, contributes importantly to Huntington's disease (HD) pathogenesis. Another neurotrophin (NT), NT-3, is reduced in the cortex of HD patients; however, its role in HD is unknown. BDNF and NT-3 bind with high affinity to the tropomyosin receptor-kinases (Trk) B and TrkC, respectively. Targeting TrkB/TrkC may be an effective HD therapeutic strategy, as multiple links exist between their signaling pathways and HD degenerative mechanisms. We developed a small molecule ligand, LM22B-10, that activates TrkB and TrkC to promote cell survival. OBJECTIVE: This study aimed to determine if upregulating TrkB/TrkC signaling with LM22B-10 would alleviate the HD phenotype in R6/2 and Q140 mice. METHODS: LM22B-10 was delivered by concomitant intranasal-intraperitoneal routes to R6/2 and Q140 mice and then motor performance and striatal pathology were evaluated. RESULTS: NT-3 levels, TrkB/TrkC phosphorylation, and AKT signaling were reduced in the R6/2 striatum; LM22B-10 counteracted these deficits. LM22B-10 also reduced intranuclear huntingtin aggregates, dendritic spine loss, microglial activation, and degeneration of dopamine- and cyclic AMP-regulated phosphoprotein with a molecular weight of 32 kDa (DARPP-32) and parvalbumin-containing neurons in the R6/2 and/or Q140 striatum. Moreover, both HD mouse models showed improved motor performance after LM22B-10 treatment. CONCLUSIONS: These results reveal an NT-3/TrkC signaling deficiency in the striatum of R6/2 mice, support the idea that targeting TrkB/TrkC alleviates HD-related neurodegeneration and motor dysfunction, and suggest a novel, disease-modifying, multi-target strategy for treating HD.

7.
bioRxiv ; 2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37781573

RESUMEN

Introduction: TrkB and TrkC receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid-ß (Aß)-toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. Methods: PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APP L/S ) and wild-type controls (WT). Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA-sequencing. Results: Memory and LTP deficits in APP L/S mice were attenuated by treatment with BD10-2. BD10-2 prevented aberrant AKT, CaMKII, and GLUA1 phosphorylation, and enhanced activity-dependent recruitment of synaptic proteins. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. Conclusions: BD10-2 prevented APP L/S /Aß-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response.

8.
Proc Natl Acad Sci U S A ; 106(12): 4906-11, 2009 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-19264961

RESUMEN

Cognitive problems occur in asymptomatic gene carriers of Huntington's disease (HD), and mouse models of the disease exhibit impaired learning and substantial deficits in the cytoskeletal changes that stabilize long-term potentiation (LTP). The latter effects may be related to the decreased production of brain-derived neurotrophic factor (BDNF) associated with the HD mutation. This study asked whether up-regulating endogenous BDNF levels with an ampakine, a positive modulator of AMPA-type glutamate receptors, rescues plasticity and reduces learning problems in HD (CAG140) mice. Twice-daily injections of a short half-life ampakine normalized BDNF levels, activity-driven actin polymerization in dendritic spines, and LTP stabilization in 8-week-old mutants. Comparable results were obtained in 16-week-old HD mice with more severe LTP deficits. Ampakine treatments had no measurable effect on the decreased locomotor activity observed in the mutants but offset their impairments in long-term memory. Given that ampakines are well tolerated in clinical trials and were effective in this study after brief exposures, these results suggest a novel strategy for chronic treatment of the cognitive difficulties that occur in the early stages of HD.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedad de Huntington/fisiopatología , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Actinas/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Animales , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Técnicas de Sustitución del Gen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Enfermedad de Huntington/complicaciones , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Trastornos de la Memoria/complicaciones , Ratones , Actividad Motora/efectos de los fármacos , Factores de Tiempo
9.
Neurobiol Dis ; 41(2): 436-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20977939

RESUMEN

Daily, systemic injections of a positive AMPA-type glutamate receptor modulator (ampakine) have been shown to reduce synaptic plasticity defects in rodent models of aging and early-stage Huntington's disease (HD). Here we report that long-term ampakine treatment markedly slows the progression of striatal neuropathology and locomotor dysfunction in the R6/2 HD mouse model. Remarkably, these effects were produced by an ampakine, CX929, with a short half-life. Injected once daily for 4-7 weeks, the compound increased protein levels of brain-derived neurotrophic factor (BDNF) in the neocortex and striatum of R6/2 but not wild-type mice. Moreover, ampakine treatments prevented the decrease in total striatal area, blocked the loss of striatal DARPP-32 immunoreactivity and reduced by 36% the size of intra-nuclear huntingtin aggregates in R6/2 striatum. The CX929 treatments also markedly improved motor performance of R6/2 mice on several measures (rotarod, vertical pole descent) but did not influence body weight or lifespan. These findings describe a minimally invasive, pharmacologically plausible strategy for treatment of HD and, potentially, other neuropathological diseases.


Asunto(s)
Antidiscinéticos/farmacología , Cuerpo Estriado/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Enfermedad de Huntington/tratamiento farmacológico , Fenotipo , Receptores AMPA/agonistas , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología , Animales , Antidiscinéticos/química , Antidiscinéticos/uso terapéutico , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Agonistas de Aminoácidos Excitadores/uso terapéutico , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Transgénicos , Receptores AMPA/fisiología , Resultado del Tratamiento
10.
Neurotherapeutics ; 18(2): 1039-1063, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33786806

RESUMEN

Huntington's disease (HD) is caused by an expansion of the CAG repeat in the huntingtin gene leading to preferential neurodegeneration of the striatum. Disease-modifying treatments are not yet available to HD patients and their development would be facilitated by translatable pharmacodynamic biomarkers. Multi-modal magnetic resonance imaging (MRI) and plasma cytokines have been suggested as disease onset/progression biomarkers, but their ability to detect treatment efficacy is understudied. This study used the R6/2 mouse model of HD to assess if structural neuroimaging and biofluid assays can detect treatment response using as a prototype the small molecule p75NTR ligand LM11A-31, shown previously to reduce HD phenotypes in these mice. LM11A-31 alleviated volume reductions in multiple brain regions, including striatum, of vehicle-treated R6/2 mice relative to wild-types (WTs), as assessed with in vivo MRI. LM11A-31 also normalized changes in diffusion tensor imaging (DTI) metrics and diminished increases in certain plasma cytokine levels, including tumor necrosis factor-alpha and interleukin-6, in R6/2 mice. Finally, R6/2-vehicle mice had increased urinary levels of the p75NTR extracellular domain (ecd), a cleavage product released with pro-apoptotic ligand binding that detects the progression of other neurodegenerative diseases; LM11A-31 reduced this increase. These results are the first to show that urinary p75NTR-ecd levels are elevated in an HD mouse model and can be used to detect therapeutic effects. These data also indicate that multi-modal MRI and plasma cytokine levels may be effective pharmacodynamic biomarkers and that using combinations of these markers would be a viable and powerful option for clinical trials.


Asunto(s)
Enfermedad de Huntington/diagnóstico por imagen , Enfermedad de Huntington/metabolismo , Isoleucina/análogos & derivados , Morfolinas/metabolismo , Morfolinas/uso terapéutico , Neuroimagen/métodos , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Biomarcadores/sangre , Biomarcadores/orina , Estudios Transversales , Evaluación Preclínica de Medicamentos/métodos , Femenino , Enfermedad de Huntington/tratamiento farmacológico , Isoleucina/metabolismo , Isoleucina/farmacología , Isoleucina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos CBA , Ratones Transgénicos , Morfolinas/farmacología
11.
Mol Cell Neurosci ; 39(1): 8-20, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18602275

RESUMEN

Huntington's disease (HD) is a devastating neurodegenerative disorder caused by an expanded polyglutamine repeat within the protein Huntingtin (Htt). We previously reported that mutant Htt expression activates the ERK1/2 and JNK pathways [Apostol, B.L., Illes, K., Pallos, J., Bodai, L., Wu, J., Strand, A., Schweitzer, E.S., Olson, J.M., Kazantsev, A., Marsh, J.L., Thompson, L.M., 2006. Mutant huntingtin alters MAPK signaling pathways in PC12 and striatal cells: ERK1/2 protects against mutant huntingtin-associated toxicity. Hum. Mol. Genet. 15, 273-285]. Chemical and genetic modulation of these pathways promotes cell survival and death, respectively. Here we test the ability of two closely related compounds, CEP-11004 and CEP-1347, which inhibit Mixed Lineage Kinases (MLKs) and are neuroprotective, to suppress mutant Htt-mediated pathogenesis in multiple model systems. CEP-11004/CEP-1347 treatment significantly decreased toxicity in mutant Htt-expressing cells that evoke a strong JNK response. However, suppression of cellular dysfunction in cell lines that exhibit only mild Htt-associated toxicity and little JNK activation was associated with activation of ERK1/2. These compounds also reduced neurotoxicity in immortalized striatal neurons from mutant knock-in mice and Drosophila expressing a mutant Htt fragment. Finally, CEP-1347 improved motor performance in R6/2 mice and restored expression of BDNF, a critical neurotrophic factor that is reduced in HD. These studies suggest a novel therapeutic approach for a currently untreatable neurodegenerative disease, HD, via CEP-1347 up-regulation of BDNF.


Asunto(s)
Animales Modificados Genéticamente , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Carbazoles/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/toxicidad , Fármacos Neuroprotectores/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/toxicidad , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Carbazoles/química , Carbazoles/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Drosophila melanogaster , Activación Enzimática , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Estructura Molecular , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/uso terapéutico , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/uso terapéutico , Proteínas Nucleares/metabolismo , Proteínas Nucleares/uso terapéutico , Fenotipo , Ratas
12.
Mol Cell Neurosci ; 39(1): 1-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18571429

RESUMEN

Huntington's disease (HD) is an age-related neurodegenerative disorder that is currently untreatable. A prominent feature of HD pathology is the reduction of the pro-survival neurotrophin Brain-Derived Neurotrophic Factor (BDNF). Both mRNA and protein levels of BDNF are decreased in the brains of several HD rodent models and in human HD patients. We now report for the first time that this molecular event is mirrored in blood from HD rodent models. While protein levels of BDNF are undetectable in mouse blood, mRNA levels are measurable and diminish during HD progression in transgenic mouse (R6/2) and rat models of HD. Among the eight different BDNF transcripts, only BDNF exon III is transcribed in mouse blood and its expression is progressively compromised in R6/2 mice with respect to age-matched wild-types. Assessment of BDNF mRNA in HD rat blood shows a similar result, which is reinforced by evidence that protein levels of the neurotrophin are also significantly reduced at a symptomatic stage. Finally, we demonstrate that acute and chronic treatment of R6/2 mice with CEP-1347, a mixed lineage kinase (MLK) inhibitor with neuroprotective and neurotrophic effects, leads to increased total BDNF mRNA in blood when compared to untreated R6/2 mice. Our results indicate that alterations in BDNF mRNA levels in peripheral blood are a readily accessible measurement of disease progression and drug efficacy in HD rodent models.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Carbazoles/metabolismo , Enfermedad de Huntington/sangre , Enfermedad de Huntington/genética , Fármacos Neuroprotectores/metabolismo , ARN Mensajero/sangre , Animales , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/genética , Carbazoles/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Exones , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/uso terapéutico , Ratas
13.
Radiother Oncol ; 139: 4-10, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31253467

RESUMEN

AIM: To evaluate the impact of ultra-rapid FLASH mouse whole brain irradiation on hippocampal dendritic spines and neuroinflammation, factors associated with cognitive impairment after brain irradiation. METHODS: We administered 30 Gy whole brain irradiation to C57BL6/J mice in sub-second (FLASH) vs. 240 s conventional delivery time keeping all other parameters constant, using a custom configured clinical linac. Ten weeks post-irradiation, we evaluated spatial and non-spatial object recognition using novel object location and object recognition testing. We measured dendritic spine density by tracing Golgi-stained hippocampal neurons and evaluated neuroinflammation by CD68 immunostaining, a marker of activated microglia, and expression of 10 pro-inflammatory cytokines using a multiplex immunoassay. RESULTS: At ten weeks post-irradiation, compared to unirradiated controls, conventional delivery time irradiation significantly impaired novel object location and recognition tasks whereas the same dose given in FLASH delivery did not. Conventional delivery time, but not FLASH, was associated with significant loss of dendritic spine density in hippocampal apical dendrites, with a similar non-significant trend in basal dendrites. Conventional delivery time was associated with significantly increased CD68-positive microglia compared to controls whereas FLASH was not. Conventional delivery time was associated with significant increases in 5 of 10 pro-inflammatory cytokines in the hippocampus (and non-significant increases in another 3), whereas FLASH was associated with smaller increases in only 3. CONCLUSION: Reduced cognitive impairment and associated neurodegeneration were observed with FLASH compared to conventional delivery time irradiation, potentially through decreased induction of neuroinflammation, suggesting a promising approach to increasing therapeutic index in radiation therapy of brain tumors.


Asunto(s)
Disfunción Cognitiva/prevención & control , Irradiación Craneana , Espinas Dendríticas/efectos de la radiación , Hipocampo/efectos de la radiación , Inflamación/prevención & control , Animales , Espinas Dendríticas/patología , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Dosificación Radioterapéutica
14.
J Neurosci ; 27(16): 4424-34, 2007 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-17442827

RESUMEN

Asymptomatic Huntington's disease (HD) patients exhibit memory and cognition deficits that generally worsen with age. Similarly, long-term potentiation (LTP), a form of synaptic plasticity involved in memory encoding, is impaired in HD mouse models well before motor disturbances occur. The reasons why LTP deteriorates are unknown. Here we show that LTP is impaired in hippocampal slices from presymptomatic Hdh(Q92) and Hdh(Q111) knock-in mice, describe two factors contributing to this deficit, and establish that potentiation can be rescued with brain-derived neurotrophic factor (BDNF). Baseline physiological measures were unaffected by the HD mutation, but LTP induction and, to a greater degree, consolidation were both defective. The facilitation of burst responses that normally occurs during a theta stimulation train was reduced in HD knock-in mice, as was theta-induced actin polymerization in dendritic spines. The decrease in actin polymerization and deficits in LTP stabilization were reversed by BDNF, concentrations of which were substantially reduced in hippocampus of both Hdh(Q92) and Hdh(Q111) mice. These results suggest that the HD mutation discretely disrupts processes needed to both induce and stabilize LTP, with the latter effect likely arising from reduced BDNF expression. That BDNF rescues LTP in HD knock-in mice suggests the possibility of treating cognitive deficits in asymptomatic HD gene carriers by upregulating production of the neurotrophin.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedad de Huntington/genética , Potenciación a Largo Plazo/genética , Plasticidad Neuronal/genética , Actinas/metabolismo , Animales , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Sinapsis/metabolismo , Ritmo Teta
15.
J Neurosci ; 27(30): 8031-9, 2007 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-17652593

RESUMEN

Stabilization of long-term potentiation (LTP) depends on multiple signaling cascades linked to actin polymerization. We used one of these, involving phosphorylation of the regulatory protein cofilin, as a marker to test whether LTP-related changes occur in hippocampal synapses during unsupervised learning. Well handled rats were allowed to explore a compartmentalized environment for 30 min after an injection of vehicle or the NMDA receptor antagonist (+/-)-3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP). Another group of rats consisted of vehicle-injected, home-cage controls. Vehicle-treated rats that explored the environment had 30% more spines with dense phosphorylated (p) cofilin immunoreactivity in hippocampal field CA1 than did rats in the home-cage group. The increase in pCofilin-positive spines and behavioral evidence for memory of the explored environment were both eliminated by CPP. Coimmunostaining for pCofilin and the postsynaptic density protein 95 (PSD-95) showed that synapses on pCofilin-positive spines were substantially larger than those on neighboring (pCofilin-negative) spines. These results establish that uncommon cellular events associated with LTP, including changes in synapse size, occur in individual spines during learning, and provide a technique for mapping potential engrams.


Asunto(s)
Hipocampo/fisiología , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Sinapsis/fisiología , Animales , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/fisiología , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos
16.
J Huntingtons Dis ; 6(4): 303-325, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29254102

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG repeat expansions in the IT15 gene which encodes the huntingtin (HTT) protein. Currently, no treatments capable of preventing or slowing disease progression exist. Disease modifying therapeutics for HD would be expected to target a comprehensive set of degenerative processes given the diverse mechanisms contributing to HD pathogenesis including neuroinflammation, excitotoxicity, and transcription dysregulation. A major contributor to HD-related degeneration is mutant HTT-induced loss of neurotrophic support. Thus, neurotrophin (NT) receptors have emerged as therapeutic targets in HD. The considerable overlap between NT signaling networks and those dysregulated by mutant HTT provides strong theoretical support for this approach. This review will focus on the contributions of disrupted NT signaling in HD-related neurodegeneration and how targeting NT receptors to augment pro-survival signaling and/or to inhibit degenerative signaling may combat HD pathologies. Therapeutic strategies involving NT delivery, peptidomimetics, and the targeting of specific NT receptors (e.g., Trks or p75NTR), particularly with small molecule ligands, are discussed.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Humanos , Transducción de Señal/efectos de los fármacos
17.
CNS Neurol Disord Drug Targets ; 16(3): 291-302, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27823570

RESUMEN

Effective non-genetic disease modifying treatments for Huntington's disease (HD) will necessarily target multiple diverse neurodegenerative processes triggered by mutant huntingtin. Neurotrophin receptors are well-positioned for this task as they regulate signaling pathways that largely overlap with signaling networks contributing to HD-related synaptic dysfunction, glial activation, excitotoxicity, and other degenerative processes. This review will discuss the contributions of disrupted neurotrophin receptor-related signaling to primary HD neuropathologies, and prospects for harnessing this signaling to develop therapeutics to counteract HD degenerative mechanisms. Application of the native protein ligands has been challenging pharmacologically, but progress has been made with the advent of small molecule compounds that can selectively bind to and activate specific Trk receptors or p75NTR to promote trophic and/or inhibit degenerative signaling in cell populations preferentially affected in HD.


Asunto(s)
Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Ligandos , Transducción de Señal/efectos de los fármacos
18.
Neuropharmacology ; 110(Pt A): 343-361, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27334657

RESUMEN

Neurotrophin (NT) receptors are coupled to numerous signaling networks that play critical roles in neuronal survival and plasticity. Several non-peptide small molecule ligands have recently been reported that bind to and activate specific tropomyosin-receptor kinase (Trk) NT receptors, stimulate their downstream signaling, and cause biologic effects similar to, though not completely overlapping, those of the native NT ligands. Here, in silico screening, coupled with low-throughput neuronal survival screening, identified a compound, LM22B-10, that, unlike prior small molecule Trk ligands, binds to and activates TrkB as well as TrkC. LM22B-10 increased cell survival and strongly accelerated neurite outgrowth, superseding the effects of brain-derived neurotrophic factor (BDNF), NT-3 or the two combined. Additionally, unlike the NTs, LM22B-10 supported substantial early neurite outgrowth in the presence of inhibiting glycoproteins. Examination of the mechanisms of these actions suggested contributions of the activation of both Trks and differential interactions with p75(NTR), as well as a requirement for involvement of the Trk extracellular domain. In aged mice, LM22B-10 activated hippocampal and striatal TrkB and TrkC, and their downstream signaling, and increased hippocampal dendritic spine density. Thus, LM22B-10 may constitute a new tool for the study of TrkB and TrkC signaling and their interactions with p75(NTR), and provides groundwork for the development of ligands that stimulate unique combinations of Trk receptors and activity patterns for application to selected neuronal populations and deficits present in various disease states.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Supervivencia Celular/fisiología , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Células HEK293 , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Proyección Neuronal/fisiología , Neuronas/citología , Neuronas/metabolismo , Ratas , Receptor trkB/agonistas , Receptor trkB/genética , Receptor trkB/metabolismo , Receptor trkC/agonistas , Receptor trkC/genética , Receptor trkC/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
19.
J Comp Neurol ; 459(3): 290-300, 2003 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-12655511

RESUMEN

The posterodorsal medial amygdala (MeApd), the posterodorsal preoptic nucleus (PdPN), and the medial cell group of the sexually dimorphic preoptic area (mSDA) contain cells that are activated specifically at ejaculation as assessed by Fos expression. The mSDA also expresses Fos early in the mating context. Because little is known about the neurotransmitters of these activated cells, the possibility that they use gamma-aminobutyric acid (GABA) or glutamate was assessed. Putative glutamatergic cells were visualized with immunocytochemistry (ICC) for glutamate and its neuron-specific transporter. Their distributions were compared with those of GABAergic cells visualized with ICC for the 67-kDa form of glutamic acid decarboxylase (GAD(67)) and in situ hybridization for GAD(67) messenger RNA (mRNA). Colocalization of Fos and GAD(67) mRNA in recently mated males indicated that half of the activated cells in the PdPN, mSDA, and lateral MeApd are GABAergic. Colocalization of Fos and glutamate suggested that a quarter of the activated mSDA and lateral MeApd cells are glutamatergic. The PdPN does not appear to have glutamatergic cells. In the lateral MeApd, the percentage of activated cells that are GABAergic (45%) matches the percentage that project to the principal part of the bed nucleus of the stria terminalis (BST; 43%), and the percentage likely to be glutamatergic (27%) matches the percentage projecting to the mSDA (27%). The latter could help to trigger ejaculation. The distribution of GABAergic and putative glutamatergic cells in the caudal preoptic area, caudal BST, and medial amygdala of male gerbils is also described.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ácido Glutámico/metabolismo , Área Preóptica/metabolismo , Conducta Sexual Animal/fisiología , Ácido gamma-Aminobutírico/metabolismo , Amígdala del Cerebelo/química , Amígdala del Cerebelo/citología , Animales , Femenino , Gerbillinae , Ácido Glutámico/análisis , Masculino , Área Preóptica/química , Área Preóptica/citología , Ácido gamma-Aminobutírico/análisis
20.
J Comp Neurol ; 444(1): 75-94, 2002 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-11835183

RESUMEN

The posterodorsal preoptic nucleus (PdPN) and the lateral part of the posterodorsal medial amygdala (MeApd) express Fos with ejaculation in male gerbils. Ejaculation-activated cells participate in the PdPN and MeApd projections to each other and to the sexually dimorphic preoptic area (SDA), but those projections involve less than 20% of the activated PdPN cells and less than 50% of the activated MeApd cells. To identify other potential targets of ejaculation-activated cells, we traced PdPN and lateral MeApd outputs using biotinylated dextran amine. The principal part of the bed nucleus of the stria terminalis (BSTpr) and the anteroventral periventricular nucleus (AVPv) were labeled from both sites and were injected with Fluoro-Gold to determine whether PdPN and lateral MeApd cells that express Fos with ejaculation would be retrogradely labeled. Fluoro-Gold was also applied to the dorsomedial hypothalamus (DMH) and retrorubral field (RRF) because such injections label PdPN cells in rats. The PdPN-DMH projection is minimal in gerbils, involving few, if any, ejaculation-related cells. Ejaculation-activated PdPN cells project to the AVPv (43%), dorsal BSTpr (30%), and RRF (12%). Those in the lateral MeApd project to the dorsal BSTpr (43%) and AVPv (18%). When these percentages are combined with those for ejaculation-activated cells involved in the PdPN and lateral MeApd projections to each other and to the medial SDA, the totals reach 100%. Thus, every PdPN and MeApd cell activated with ejaculation may participate in one of these projections. Similar projections may contribute to the similar behavioral effects of the PdPN and MeApd.


Asunto(s)
Amígdala del Cerebelo/citología , Biotina/análogos & derivados , Eyaculación/fisiología , Gerbillinae/anatomía & histología , Área Preóptica/citología , Estilbamidinas , Vías Aferentes , Amígdala del Cerebelo/fisiología , Animales , Dextranos , Núcleo Hipotalámico Dorsomedial/citología , Núcleo Hipotalámico Dorsomedial/fisiología , Vías Eferentes , Femenino , Colorantes Fluorescentes , Masculino , Área Preóptica/fisiología , Proteínas Proto-Oncogénicas c-fos/análisis , Caracteres Sexuales , Tegmento Mesencefálico/citología , Tegmento Mesencefálico/fisiología , Tirosina 3-Monooxigenasa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA