Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Pharmacol Transl Sci ; 4(1): 143-154, 2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33615168

RESUMEN

Selective liver X receptor (LXR) agonists have been extensively pursued as therapeutics for Alzheimer's disease and related dementia (ADRD) and, for comorbidities such as type 2 diabetes (T2D) and cerebrovascular disease (CVD), disorders with underlying impaired insulin signaling, glucose metabolism, and cholesterol mobilization. The failure of the LXR-focused approach led us to pursue a novel strategy to discover nonlipogenic ATP-binding cassette transporter A1 (ABCA1) inducers (NLAIs): screening for ABCA1-luciferase activation in astrocytoma cells and counterscreening against lipogenic gene upregulation in hepatocarcinoma cells. Beneficial effects of LXRß agonists mediated by ABCA1 include the following: control of cholesterol and phospholipid efflux to lipid-poor apolipoproteins forming beneficial peripheral HDL and HDL-like particles in the brain and attenuation of inflammation. While rare, ABCA1 variants reduce plasma HDL and correlate with an increased risk of ADRD and CVD. In secondary assays, NLAI hits enhanced cholesterol mobilization and positively impacted in vitro biomarkers associated with insulin signaling, inflammatory response, and biogenic properties. In vivo target engagement was demonstrated after oral administration of NLAIs in (i) mice fed a high-fat diet, a model for obesity-linked T2D, (ii) mice administered LPS, and (iii) mice with accelerated oxidative stress. The lack of adverse effects on lipogenesis and positive effects on multiple biomarkers associated with T2D and ADRD supports this novel phenotypic approach to NLAIs as a platform for T2D and ADRD drug discovery.

2.
Nitric Oxide ; 82: 59-74, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30394348

RESUMEN

The development of small molecule modulators of NO/cGMP signaling for use in the CNS has lagged far behind the use of such clinical agents in the periphery, despite the central role played by NO/cGMP in learning and memory, and the substantial evidence that this signaling pathway is perturbed in neurodegenerative disorders, including Alzheimer's disease. The NO-chimeras, NMZ and Nitrosynapsin, have yielded beneficial and disease-modifying responses in multiple preclinical animal models, acting on GABAA and NMDA receptors, respectively, providing additional mechanisms of action relevant to synaptic and neuronal dysfunction. Several inhibitors of cGMP-specific phosphodiesterases (PDE) have replicated some of the actions of these NO-chimeras in the CNS. There is no evidence that nitrate tolerance is a phenomenon relevant to the CNS actions of NO-chimeras, and studies on nitroglycerin in the periphery continue to challenge the dogma of nitrate tolerance mechanisms. Hybrid nitrates have shown much promise in the periphery and CNS, but to date only one treatment has received FDA approval, for glaucoma. The potential for allosteric modulation of soluble guanylate cyclase (sGC) in brain disorders has not yet been fully explored nor exploited; whereas multiple applications of PDE inhibitors have been explored and many have stalled in clinical trials.


Asunto(s)
Fármacos del Sistema Nervioso Central/farmacología , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Enfermedades del Sistema Nervioso Central/metabolismo , GMP Cíclico/metabolismo , Descubrimiento de Drogas , Óxido Nítrico/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Fármacos del Sistema Nervioso Central/síntesis química , Fármacos del Sistema Nervioso Central/química , Humanos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
3.
BMC Res Notes ; 11(1): 662, 2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213274

RESUMEN

OBJECTIVE: The Uncoordinated 5A (UNC5A) protein is part of a family of receptors that play roles in axonal pathfinding and cell migration. We previously showed that the Fanconi anemia C protein (FANCC) interacts with UNC5A and delays UNC5A-mediated apoptosis. FANCC is a predominantly cytoplasmic protein that has multiple functions including DNA damage signaling, oxygen radical metabolism, signal transduction, transcriptional regulation and apoptosis. Given the direct interaction between FANCC and UNC5A and that FANCC interferes with UNC5A-mediated apoptosis, we explored the possibility that FANCC might play a role in axonal-like growth processes. RESULTS: Here we show that FANCC and UNC5A are localized to regions of neurite outgrowth during neuronal cell differentiation. We also show that absence of FANCC is required for neurite outgrowth. In addition, FANCC seems required for UNC5A expression. Results from this study combined with our previous report suggest that FANCC plays a role in tissue development through the regulation of UNC5A-mediated functions.


Asunto(s)
Proteína del Grupo de Complementación C de la Anemia de Fanconi/fisiología , Receptores de Netrina/fisiología , Proyección Neuronal , Animales , Diferenciación Celular , Anemia de Fanconi , Ratones , Ratones Noqueados , Proteínas
4.
J Biol Chem ; 293(41): 16069-16082, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30154245

RESUMEN

Prion diseases are fatal infectious neurodegenerative disorders in humans and other animals and are caused by misfolding of the cellular prion protein (PrPC) into the pathological isoform PrPSc These diseases have the potential to transmit within or between species, including zoonotic transmission to humans. Elucidating the molecular and cellular mechanisms underlying prion propagation and transmission is therefore critical for developing molecular strategies for disease intervention. We have shown previously that impaired quality control mechanisms directly influence prion propagation. In this study, we manipulated cellular quality control pathways in vitro by stably and transiently overexpressing selected quality control folding (ERp57) and cargo (VIP36) proteins and investigated the effects of this overexpression on prion propagation. We found that ERp57 or VIP36 overexpression in persistently prion-infected neuroblastoma cells significantly reduces the amount of PrPSc in immunoblots and prion-seeding activity in the real-time quaking-induced conversion (RT-QuIC) assay. Using different cell lines infected with various prion strains confirmed that this effect is not cell type- or prion strain-specific. Moreover, de novo prion infection revealed that the overexpression significantly reduced newly formed PrPSc in acutely infected cells. ERp57-overexpressing cells significantly overcame endoplasmic reticulum stress, as revealed by expression of lower levels of the stress markers BiP and CHOP, accompanied by a decrease in PrP aggregates. Furthermore, application of ERp57-expressing lentiviruses prolonged the survival of prion-infected mice. Taken together, improved cellular quality control via ERp57 or VIP36 overexpression impairs prion propagation and could be utilized as a potential therapeutic strategy.


Asunto(s)
Lectinas de Unión a Manosa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Animales , Línea Celular Tumoral , Estrés del Retículo Endoplásmico , Femenino , Expresión Génica , Humanos , Lectinas de Unión a Manosa/genética , Proteínas de Transporte de Membrana/genética , Ratones , Proteína Disulfuro Isomerasas/genética
5.
ACS Chem Neurosci ; 9(5): 894-900, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29338172

RESUMEN

Activation of HIF-1α and Nrf2 is a primary component of cellular response to oxidative stress, and activation of HIF-1α and Nrf2 provides neuroprotection in models of neurodegenerative disorders, including ischemic stroke, Alzheimer's and Parkinson's diseases. Screening a library of CNS-targeted drugs using novel reporters for HIF-1α and Nrf2 elevation in neuronal cells revealed histone deacetylase (HDAC) inhibitors as potential activators of these pathways. We report the identification of phenylhydroxamates as single agents exhibiting tripartite inhibition of HDAC6, inhibition of HIF-1 prolyl hydroxylase (PHD), and activation of Nrf2. Two superior tripartite agents, ING-6 and ING-66, showed neuroprotection against various cellular insults, associated with stabilization of both Nrf2 and HIF-1, and expression of their respective target genes in vitro and in vivo. Discovery of the innate ability of phenylhydroxamate HDAC inhibitors to activate Nrf2 and HIF provides a novel route to multifunctional neuroprotective agents and cautions against HDAC6 selective inhibitors as chemical probes of specific HDAC isoform function.


Asunto(s)
Histona Desacetilasa 6/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Hidroxilaminas/farmacología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
6.
Cephalalgia ; 38(8): 1471-1484, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29022756

RESUMEN

Background Nitric oxide (NO) has been heavily implicated in migraine. Nitroglycerin is a prototypic NO-donor, and triggers migraine in humans. However, nitroglycerin also induces oxidative/nitrosative stress and is a source of peroxynitrite - factors previously linked with migraine etiology. Soluble guanylyl cyclase (sGC) is the high affinity NO receptor in the body, and the aim of this study was to identify the precise role of sGC in acute and chronic migraine. Methods We developed a novel brain-bioavailable sGC stimulator (VL-102), and tested its hyperalgesic properties in mice. We also determined the effect of VL-102 on c-fos and calcitonin gene related peptide (CGRP) immunoreactivity within the trigeminovascular complex. In addition, we also tested the known sGC inhibitor, ODQ, within the chronic nitroglycerin migraine model. Results VL-102-evoked acute and chronic mechanical cephalic and hind-paw allodynia in a dose-dependent manner, which was blocked by the migraine medications sumatriptan, propranolol, and topiramate. In addition, VL-102 also increased c-fos and CGRP expressing cells within the trigeminovascular complex. Importantly, ODQ completely inhibited acute and chronic hyperalgesia induced by nitroglycerin. ODQ also blocked hyperalgesia already established by chronic nitroglycerin, implicating this pathway in migraine chronicity. Conclusions These results indicate that nitroglycerin causes migraine-related pain through stimulation of the sGC pathway, and that super-activation of this receptor may be an important component for the maintenance of chronic migraine. This work opens the possibility for negative sGC modulators as novel migraine therapies.


Asunto(s)
Hiperalgesia/inducido químicamente , Hiperalgesia/enzimología , Trastornos Migrañosos/enzimología , Guanilil Ciclasa Soluble/fisiología , Antagonistas Adrenérgicos beta/administración & dosificación , Antagonistas Adrenérgicos beta/uso terapéutico , Regulación Alostérica , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/uso terapéutico , Péptido Relacionado con Gen de Calcitonina/biosíntesis , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Femenino , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/etiología , Terapia Molecular Dirigida , Óxido Nítrico/efectos adversos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Nitroglicerina/farmacología , Oxadiazoles/administración & dosificación , Oxadiazoles/metabolismo , Oxadiazoles/uso terapéutico , Propranolol/administración & dosificación , Propranolol/uso terapéutico , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Quinoxalinas/administración & dosificación , Quinoxalinas/metabolismo , Quinoxalinas/uso terapéutico , Agonistas del Receptor de Serotonina 5-HT1/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT1/uso terapéutico , Guanilil Ciclasa Soluble/metabolismo , Sumatriptán/administración & dosificación , Sumatriptán/uso terapéutico , Topiramato/administración & dosificación , Topiramato/uso terapéutico
7.
Curr Med Chem ; 23(24): 2770-2788, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27776476

RESUMEN

cAMP-response element-binding protein (CREB) plays a central role in various aspects of central nervous system (CNS) function, ranging from the developmental stages to neuronal plasticity and survival in adult brain. Activation of CREB plays a crucial role in learning and memory and is at the convergence of multiple intracellular signaling cascades including CAMKII and MAPK. This review focuses on the important functions of nitric oxide (NO) in activating CREB via the NO receptor, soluble guanylyl cyclase (sGC), and production of the second messenger, cGMP. The involvement of the NO/cGMP signaling pathway in synaptic plasticity suggests several avenues for therapeutic intervention, and targeting early synaptic degeneration could be an attractive approach for the development of novel disease-modifying approaches to treat cognition and memory dysfunction in neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/patología , Sistema Nervioso Central/metabolismo , GMP Cíclico/metabolismo , Enfermedades Neurodegenerativas/patología , Óxido Nítrico/metabolismo , Enfermedad de Alzheimer/metabolismo , Proteína de Unión a CREB/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Plasticidad Neuronal/fisiología , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/metabolismo , Transducción de Señal , Guanilil Ciclasa Soluble/antagonistas & inhibidores , Guanilil Ciclasa Soluble/metabolismo , Sinapsis/metabolismo
8.
Mol Neurodegener ; 11: 35, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27129593

RESUMEN

BACKGROUND: Clinical failures singularly targeting amyloid-ß pathology indicate a critical need for alternative Alzheimer's disease (AD) therapeutic strategies. The mixed pathology reported in a large population of AD patients demands a multifunctional drug approach. Since activation of cAMP response element binding protein (CREB) plays a crucial role in synaptic strengthening and memory formation, we retooled a clinical drug with known neuroprotective and anti-inflammatory activity to activate CREB, and validated this novel multifunctional drug, NMZ, in 4 different mouse models of AD. RESULTS: NMZ was tested in three mouse models of familial AD and one model of sporadic AD. In 3 × Tg hippocampal slices, NMZ restored LTP. In vivo, memory was improved with NMZ in all animal models with robust cognitive deficits. NMZ treatment lowered neurotoxic forms of Aß in both APP/PS1 and 3 × Tg transgenic mice while also restoring neuronal plasticity biomarkers in the 3 × Tg mice. In EFAD mice, incorporation of the major genetic AD risk factor, hAPOE4, did not mute the beneficial drug effects. In a novel sporadic mouse model that manifests AD-like pathology caused by accelerated oxidative stress in the absence of any familial AD mutation, oral administration of NMZ attenuated hallmark AD pathology and restored biomarkers of synaptic and neuronal function. CONCLUSIONS: The multifunctional approach, embodied by NMZ, was successful in mouse models of AD incorporating Aß pathology (APP/PS1), tau pathology (3xTg), and APOE4, the major human genetic risk factor for AD (EFAD). The efficacy observed in a novel model of sporadic AD (Aldh2 (-/-) ) demonstrates that the therapeutic approach is not limited to rare, familial AD genetic mutations. The multifunctional drug, NMZ, was not designed directly to target Aß and tau pathology; however, the attenuation of this hallmark pathology suggests the approach to be a highly promising, disease-modifying strategy for AD and mixed pathology dementia.


Asunto(s)
Enfermedad de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Trastornos del Conocimiento/metabolismo , Memoria/fisiología , Plasticidad Neuronal/fisiología , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones Noqueados , Ratones Transgénicos
9.
BMC Neurosci ; 16: 67, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26480871

RESUMEN

BACKGROUND: Synaptic dysfunction is a key event in pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD) where synapse loss pathologically correlates with cognitive decline and dementia. Although evidence suggests that aberrant protein production and aggregation are the causative factors in familial subsets of such diseases, drugs singularly targeting these hallmark proteins, such as amyloid-ß, have failed in late stage clinical trials. Therefore, to provide a successful disease-modifying compound and address synaptic dysfunction and memory loss in AD and mixed pathology dementia, we repurposed a clinically proven drug, CMZ, with neuroprotective and anti-inflammatory properties via addition of nitric oxide (NO) and cGMP signaling property. RESULTS: The novel compound, NMZ, was shown to retain the GABAA potentiating actions of CMZ in vitro and sedative activity in vivo. Importantly, NMZ restored LTP in hippocampal slices from AD transgenic mice, whereas CMZ was without effect. NMZ reversed amnestic blockade of acetylcholine receptors by scopolamine as well as NMDA receptor blockade by a benzodiazepine and a NO synthase inhibitor in the step-through passive avoidance (STPA) test of learning and working memory. A PK/PD relationship was developed based on STPA analysis coupled with pharmacokinetic measures of drug levels in the brain: at 1 nM concentration in brain and plasma, NMZ was able to restore memory consolidation in mice. CONCLUSION: Our findings show that NMZ embodies a promising pharmacological approach targeting synaptic dysfunction and opens new avenues for neuroprotective intervention strategies in mixed pathology AD, neurodegeneration, and dementia.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Clormetiazol/análogos & derivados , Reposicionamiento de Medicamentos/métodos , Agonistas de Receptores de GABA-A/farmacología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Animales , Proteína de Unión a CREB/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Agonistas de Receptores de GABA-A/farmacocinética , Masculino , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/farmacocinética , Óxido Nítrico/metabolismo , Nootrópicos/farmacocinética , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/patología , Xenopus laevis
10.
Mol Neurodegener ; 10: 7, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25871877

RESUMEN

BACKGROUND: Alzheimer's disease (AD) causes progressive loss of memory and cognition, exacerbated by APOE4, the greatest genetic risk factor for AD. One proposed mechanism for apolipoprotein E (apoE) effects on cognition is via NMDAR-dependent signaling. APOE genotype-specific effects on this pathway were dissected using EFAD-transgenic (Tg) mice (5xFAD mice, that over-express human amyloid-beta (Aß) via 5 familial-AD (FAD) mutations, and express human apoE), and 5xFAD/APOE-knockout (KO) mice. Previous data from EFAD-Tg mice demonstrate age-dependent (2-6 months), apoE-specific effects on the development of Aß pathology. This study tests the hypothesis that apoE4 impairs cognition via modulation of NMDAR-dependent signaling, specifically via a loss of function by comparison of E4FAD mice with 5xFAD/APOE-KO mice, E3FAD and E2FAD mice. RESULTS: Using female E2FAD, E3FAD, E4FAD and 5xFAD/APOE-KO mice aged 2-, 4-, and 6-months, the Y-maze and Morris water maze behavioral tests were combined with synaptic protein levels as markers of synaptic viability. The results demonstrate a greater age-induced deficit in cognition and reduction in PSD95, drebrin and NMDAR subunits in the E4FAD and 5xFAD/APOE-KO mice compared with E2FAD and E3FAD mice, consistent with an apoE4 loss of function. Interestingly, for NMDAR-mediated signaling, the levels of p-CaMK-II followed this same apoE-specific pattern as cognition, while the levels of p-CREB and BDNF demonstrate an apoE4 toxic gain of function: E2FAD > E3FAD > 5xFAD/APOE-KO > E4FAD. CONCLUSION: These findings suggest that compared with E2FAD and E3FAD, E4FAD and 5xFAD/APOE-KO mice exhibit enhanced age-induced reductions in cognition and key synaptic proteins via down-regulation of an NMDAR signaling pathway, consistent with an apoE4 loss of function. However, levels of p-CREB and BDNF, signaling factors common to multiple pathways, suggest a gain of toxic function. Publications in this field present contradictory results as to whether APOE4 imparts a loss or gain of function. As with the results reported herein, the overall effect of APOE4 on a given CNS-specific measure will be the product of multiple overlapping mechanisms. Thus, caution remains critical in determining whether APOE gene inactivation or therapies that correct the loss of positive function related to apoE4, are the appropriate therapeutic response.


Asunto(s)
Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Cognición/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/genética , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Regulación hacia Abajo , Memoria/fisiología , Ratones Noqueados , Mutación/genética , Transducción de Señal/fisiología
11.
J Neurochem ; 133(4): 465-88, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25689586

RESUMEN

Chronic glial activation and neuroinflammation induced by the amyloid-ß peptide (Aß) contribute to Alzheimer's disease (AD) pathology. APOE4 is the greatest AD-genetic risk factor; increasing risk up to 12-fold compared to APOE3, with APOE4-specific neuroinflammation an important component of this risk. This editorial review discusses the role of APOE in inflammation and AD, via a literature review, presentation of novel data on Aß-induced neuroinflammation, and discussion of future research directions. The complexity of chronic neuroinflammation, including multiple detrimental and beneficial effects occurring in a temporal and cell-specific manner, has resulted in conflicting functional data for virtually every inflammatory mediator. Defining a neuroinflammatory phenotype (NIP) is one way to address this issue, focusing on profiling the changes in inflammatory mediator expression during disease progression. Although many studies have shown that APOE4 induces a detrimental NIP in peripheral inflammation and Aß-independent neuroinflammation, data for APOE-modulated Aß-induced neuroinflammation are surprisingly limited. We present data supporting the hypothesis that impaired apoE4 function modulates Aß-induced effects on inflammatory receptor signaling, including amplification of detrimental (toll-like receptor 4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways. To ultimately develop APOE genotype-specific therapeutics, it is critical that future studies define the dynamic NIP profile and pathways that underlie APOE-modulated chronic neuroinflammation. In this editorial review, we present data supporting the hypothesis that impaired apoE4 function modulates Aß-induced effects on inflammatory receptor signaling, including amplification of detrimental (TLR4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways, resulting in an adverse NIP that causes neuronal dysfunction. NIP, Neuroinflammatory phenotype; P.I., pro-inflammatory; A.I., anti-inflammatory.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/toxicidad , Apolipoproteínas E/fisiología , Inflamación/etiología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Humanos , Inflamación/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Transducción de Señal/efectos de los fármacos
12.
J Biol Chem ; 289(44): 30538-30555, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25217640

RESUMEN

Previous data demonstrate that bexarotene (Bex), retinoid X receptor (RXR) agonist, reduces soluble and insoluble amyloid-ß (Aß) in Alzheimer disease (AD)-transgenic mice either by increasing the levels of mouse apolipoprotein E (apoE) or increasing ABCA1/ABCG1-induced apoE lipoprotein association/lipidation. However, although the mechanism of action of RXR agonists remains unclear, a major concern for their use is human (h)-APOE4, the greatest AD genetic risk factor. If APOE4 imparts a toxic gain-of-function, then increasing apoE4 may increase soluble Aß, likely the proximal AD neurotoxin. If the APOE4 loss-of-function is lipidation of apoE4, then induction of ABCA1/ABCG1 may be beneficial. In novel EFAD-Tg mice (overexpressing h-Aß42 with h-APOE), levels of soluble Aß (Aß42 and oligomeric Aß) are highest in E4FAD hippocampus (HP) > E3FAD-HP > E4FAD cortex (CX) > E3FAD-CX, whereas levels of lipoprotein-associated/lipidated apoE have the opposite pattern (6 months). In E4FAD-HP, short-term RXR agonist treatment (Bex or LG100268; 5.75-6 months) increased ABCA1, apoE4 lipoprotein-association/lipidation, and apoE4/Aß complex, decreased soluble Aß, and increased PSD95. In addition, hydrogel delivery, which mimics low sustained release, was equally effective as gavage for Bex and LG100268. RXR agonists induced no beneficial effects in the E4FAD-HP in a prevention protocol (5-6 months) and actually increased soluble Aß levels in E3FAD-CX and E4FAD-CX with the short-term protocol, possibly the result of systemic hepatomegaly. Thus, RXR agonists address the loss-of-function associated with APOE4 and exacerbated by Aß pathology, i.e. low levels of apoE4 lipoprotein association/lipidation. Further studies are vital to address whether RXR agonists are an APOE4-specific AD therapeutic and the systemic side effects that limit translational application.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Ácidos Nicotínicos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Receptores X Retinoide/agonistas , Tetrahidronaftalenos/administración & dosificación , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/metabolismo , Administración Oral , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Bexaroteno , Homólogo 4 de la Proteína Discs Large , Evaluación Preclínica de Medicamentos , Genotipo , Guanilato-Quinasas/metabolismo , Humanos , Lipoproteínas/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Ácidos Nicotínicos/efectos adversos , Ácidos Nicotínicos/farmacocinética , Tamaño de los Órganos/efectos de los fármacos , Receptores X Retinoide/metabolismo , Solubilidad , Tetrahidronaftalenos/efectos adversos , Tetrahidronaftalenos/farmacocinética
13.
PLoS One ; 9(3): e92811, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24676280

RESUMEN

The Fanconi anemia group C protein (FANCC) is one of the several proteins that comprise the Fanconi anemia (FA) network involved in genomic surveillance. FANCC is mainly cytoplasmic and has many functions, including apoptosis suppression through caspase-mediated proteolytic processing. Here, we examined the role of FANCC proteolytic fragments by identifying their binding partners. We performed a yeast two-hybrid screen with caspase-mediated FANCC cleavage products and identified the dependence receptor uncoordinated-5A (UNC5A) protein. Here, we show that FANCC physically interacts with UNC5A, a pro-apoptotic dependence receptor. FANCC interaction occurs through the UNC5A intracellular domain, specifically via its death domain. FANCC modulates cell sensitivity to UNC5A-mediated apoptosis; we observed reduced UNC5A-mediated apoptosis in the presence of FANCC and increased apoptosis in FANCC-depleted cells. Our results show that FANCC interferes with UNC5A's functions in apoptosis and suggest that FANCC may participate in developmental processes through association with the dependence receptor UNC5A.


Asunto(s)
Apoptosis , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Citoplasma , Humanos , Modelos Biológicos , Receptores de Netrina , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Transporte de Proteínas , Receptores de Superficie Celular/química , Técnicas del Sistema de Dos Híbridos
14.
Int J Alzheimers Dis ; 2012: 947147, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22482079

RESUMEN

Alzheimer's disease (AD) etiological studies suggest that an elevation in amyloid-ß peptides (Aß) level contributes to aggregations of the peptide and subsequent development of the disease. The major constituent of these amyloid peptides is the 1 to 40-42 residue peptide (Aß(40-42)) derived from amyloid protein precursor (APP). Most likely, reducing Aß levels in the brain may block both its aggregation and neurotoxicity and would be beneficial for patients with AD. Among the several possible ways to lower Aß accumulation in the cells, we have selectively chosen to target the primary step in the Aß cascade, namely, to reduce APP gene expression. Toward this end, we engineered specific SOFA-HDV ribozymes, a new generation of catalytic RNA tools, to decrease APP mRNA levels. Additionally, we demonstrated that APP-ribozymes are effective at decreasing APP mRNA and protein levels as well as Aß levels in neuronal cells. Our results could lay the groundwork for a new protective treatment for AD.

15.
Biochem J ; 388(Pt 2): 455-64, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15656785

RESUMEN

We have purified and characterized two peptides, named KAaH1 and KAaH2 (AaH polypeptides 1 and 2 active on K+ channels, where AaH stands for Androctonus australis Hector), from the venom of A. australis Hector scorpions. Their sequences contain 58 amino acids including six half-cysteines and differ only at positions 26 (Phe/Ser) and 29 (Lys/Gln). Although KAaH1 and KAaH2 show important sequence similarity with anti-mammal beta toxins specific for voltage-gated Na+ channels, only weak beta-like effects were observed when KAaH1 or KAaH2 (1 microM) were tested on brain Nav1.2 channels. In contrast, KAaH1 blocks Kv1.1 and Kv1.3 channels expressed in Xenopus oocytes with IC50 values of 5 and 50 nM respectively, whereas KAaH2 blocks only 20% of the current on Kv1.1 and is not active on Kv1.3 channels at a 100 nM concentration. KAaH1 is thus the first member of a new subfamily of long-chain toxins mainly active on voltage-gated K+ channels. NMR spectra of KAaH1 and KAaH2 show good dispersion of signals but broad lines and poor quality. Self-diffusion NMR experiments indicate that lines are broadened due to a conformational exchange on the millisecond time scale. NMR and CD indicate that both polypeptides adopt a similar fold with alpha-helical and b-sheet structures. Homology-based molecular models generated for KAaH1 and KAaH2 are in accordance with CD and NMR data. In the model of KAaH1, the functionally important residues Phe26 and Lys29 are close to each other and are located in the alpha-helix. These residues may constitute the so-called functional dyad observed for short alpha-KTx scorpion toxins in the beta-sheet.


Asunto(s)
Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacología , Proteínas/química , Proteínas/farmacología , Venenos de Escorpión/química , Venenos de Escorpión/farmacología , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos , Femenino , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oocitos , Unión Proteica , Conformación Proteica , Proteínas/toxicidad , Venenos de Escorpión/toxicidad , Escorpiones , Homología de Secuencia de Aminoácido , Canales de Sodio/efectos de los fármacos , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA