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.
Biomolecules ; 13(2)2023 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-36830589

RESUMEN

Misfolded aggregation of the hyperphosphorylated microtubule binding protein Tau in the brain is a pathological hallmark of Alzheimer's disease (AD). Tau aggregation downregulates brain-derived neurotrophic factor (BDNF)/tropomycin receptor kinase B (TRKB) signaling and leads to neurotoxicity. Therefore, enhancement of BDNF/TRKB signaling could be a strategy to alleviate Tau neurotoxicity. In this study, eight compounds were evaluated for the potential of inhibiting Tau misfolding in human neuroblastoma SH-SY5Y cells expressing the pro-aggregator Tau folding reporter (ΔK280 TauRD-DsRed). Among them, coumarin derivative ZN-015 and quinoline derivatives VB-030 and VB-037 displayed chemical chaperone activity to reduce ΔK280 TauRD aggregation and promote neurite outgrowth. Studies of TRKB signaling revealed that ZN-015, VB-030 and VB-037 treatments significantly increased phosphorylation of TRKB and downstream Ca2+/calmodulin-dependent protein kinase II (CaMKII), extracellular signal-regulated kinase 1/2 (ERK) and AKT serine/threonine kinase (AKT), to activate ribosomal S6 kinase (RSK) and cAMP response element-binding protein (CREB). Subsequently, p-CREB enhanced the transcription of pro-survival BDNF and BCL2 apoptosis regulator (BCL2), accompanied with reduced expression of anti-survival BCL2-associated X protein (BAX) in ΔK280 TauRD-DsRed-expressing cells. The neurite outgrowth promotion effect of ZN-015, VB-030 and VB-037 was counteracted by a RNA interference-mediated knockdown of TRKB, suggesting the role of these compounds acting as TRKB agonists. Tryptophan fluorescence quenching analysis showed that ZN-015, VB-030 and VB-037 interacted directly with a Pichia pastoris-expressed TRKB extracellular domain, indirectly supporting the role through TRKB signaling. The results of up-regulation in TRKB signaling open up the therapeutic potentials of ZN-015, VB-030 and VB-037 for AD.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Enfermedad de Alzheimer/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Proto-Oncogénicas c-akt , Neuroblastoma/metabolismo , Proteínas tau/metabolismo , Receptor trkB/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2
2.
Int J Mol Sci ; 24(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36768965

RESUMEN

Neuroinflammation and oxidative stress have been emerging as important pathways contributing to Parkinson's disease (PD) pathogenesis. In PD brains, the activated microglia release inflammatory factors such as interleukin (IL)-ß, IL-6, tumor necrosis factor (TNF)-α, and nitric oxide (NO), which increase oxidative stress and mediate neurodegeneration. Using 1-methyl-4-phenylpyridinium (MPP+)-activated human microglial HMC3 cells and the sub-chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, we found the potential of indole derivative NC009-1 against neuroinflammation, oxidative stress, and neurodegeneration for PD. In vitro, NC009-1 alleviated MPP+-induced cytotoxicity, reduced NO, IL-1ß, IL-6, and TNF-α production, and suppressed NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in MPP+-activated HMC3 cells. In vivo, NC009-1 ameliorated motor deficits and non-motor depression, increased dopamine and dopamine transporter levels in the striatum, and reduced oxidative stress as well as microglia and astrocyte reactivity in the ventral midbrain of MPTP-treated mice. These protective effects were achieved by down-regulating NLRP3, CASP1, iNOS, IL-1ß, IL-6, and TNF-α, and up-regulating SOD2, NRF2, and NQO1. These results strengthen the involvement of neuroinflammation and oxidative stress in PD pathogenic mechanism, and indicate NC009-1 as a potential drug candidate for PD treatment.


Asunto(s)
Enfermedad de Parkinson , Ratones , Humanos , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Neurotoxinas/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedades Neuroinflamatorias , Interleucina-6/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Microglía/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Estrés Oxidativo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos
3.
Biomol Ther (Seoul) ; 31(3): 285-297, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36646447

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease with progressive memory loss and the cognitive decline. AD is mainly caused by abnormal accumulation of misfolded amyloid ß (Aß), which leads to neurodegeneration via a number of possible mechanisms such as down-regulation of brain-derived neurotrophic factor-tropomyosin-related kinase B (BDNF-TRKB) signaling pathway. 7 ,8-Dihydroxyflavone (7,8-DHF), a TRKB agonist, has demonstrated potential to enhance BDNF-TRKB pathway in various neurodegenerative diseases. To expand the capacity of flavones as TRKB agonists, two natural flavones quercetin and apigenin, were evaluated. With tryptophan fluorescence quenching assay, we illustrated the direct interaction between quercetin/apigenin and TRKB extracellular domain. Employing Aß folding reporter SH-SY5Y cells, we showed that quercetin and apigenin reduced Aß-aggregation, oxidative stress, caspase-1 and acetylcholinesterase activities, as well as improved the neurite outgrowth. Treatments with quercetin and apigenin increased TRKB Tyr516 and Tyr817 and downstream cAMP-response-element binding protein (CREB) Ser133 to activate transcription of BDNF and BCL2 apoptosis regulator (BCL2), as well as reduced the expression of pro-apoptotic BCL2 associated X protein (BAX). Knockdown of TRKB counteracted the improvement of neurite outgrowth by quercetin and apigenin. Our results demonstrate that quercetin and apigenin are to work likely as a direct agonist on TRKB for their neuroprotective action, strengthening the therapeutic potential of quercetin and apigenin in treating AD.

4.
Int J Mol Sci ; 23(21)2022 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-36361524

RESUMEN

Hyperphosphorylation and aggregation of the microtubule binding protein tau is a neuropathological hallmark of Alzheimer's disease/tauopathies. Tau neurotoxicity provokes alterations in brain-derived neurotrophic factor (BDNF)/tropomycin receptor kinase B (TRKB)/cAMP-response-element binding protein (CREB) signaling to contribute to neurodegeneration. Compounds activating TRKB may therefore provide beneficial effects in tauopathies. LM-031, a coumarin derivative, has demonstrated the potential to improve BDNF signaling in neuronal cells expressing pro-aggregated ΔK280 tau mutant. In this study, we investigated if LM-031 analogous compounds provide neuroprotection effects through interaction with TRKB in SH-SY5Y cells expressing ΔK280 tauRD-DsRed folding reporter. All four LMDS compounds reduced tau aggregation and reactive oxygen species. Among them, LMDS-1 and -2 reduced caspase-1, caspase-6 and caspase-3 activities and promoted neurite outgrowth, and the effect was significantly reversed by knockdown of TRKB. Treatment of ERK inhibitor U0126 or PI3K inhibitor wortmannin decreased p-CREB, BDNF and BCL2 in these cells, implying that the neuroprotective effects of LMDS-1/2 are via activating TRKB downstream ERK, PI3K-AKT and CREB signaling. Furthermore, LMDS-1/2 demonstrated their ability to quench the intrinsic fluorescence of tryptophan residues within the extracellular domain of TRKB, thereby consolidating their interaction with TRKB. Our results suggest that LMDS-1/2 exert neuroprotection through activating TRKB signaling, and shed light on their potential application in therapeutics of Alzheimer's disease/tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Fármacos Neuroprotectores , Tauopatías , Humanos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas tau/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neuroprotección , Enfermedad de Alzheimer/tratamiento farmacológico , Cumarinas/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Neuroblastoma/metabolismo , Receptor trkB/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Caspasas
5.
Aging (Albany NY) ; 14(18): 7568-7586, 2022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-36170028

RESUMEN

Decreased BDNF and impaired TRKB signaling contribute to neurodegeneration in Alzheimer's disease (AD). We have shown previously that coumarin derivative LM-031 enhanced CREB/BDNF/BCL2 pathway. In this study we explored if LM-031 analogs LMDS-1 to -4 may act as TRKB agonists to protect SH-SY5Y cells against Aß toxicity. By docking computation for binding with TRKB using 7,8-DHF as a control, all four LMDS compounds displayed potential of binding to domain d5 of TRKB. In addition, all four LMDS compounds exhibited anti-aggregation and neuroprotective efficacy on SH-SY5Y cells with induced Aß-GFP expression. Knock-down of TRKB significantly attenuated TRKB downstream signaling and the neurite outgrowth-promoting effects of these LMDS compounds. Among them, LMDS-1 and -2 were further examined for TRKB signaling. Treatment of ERK inhibitor U0126 or PI3K inhibitor wortmannin decreased p-CREB, BDNF and BCL2 in Aß-GFP cells, implicating the neuroprotective effects are via activating TRKB downstream ERK, PI3K-AKT and CREB signaling. LMDS-1 and -2 are blood-brain barrier permeable as shown by parallel artificial membrane permeability assay. Our results demonstrate how LMDS-1 and -2 are likely to work as TRKB agonists to exert neuroprotection in Aß cells, which may shed light on the potential application in therapeutics of AD.


Asunto(s)
Enfermedad de Alzheimer , Glicoproteínas de Membrana/agonistas , Neuroblastoma , Fármacos Neuroprotectores , Receptor trkB/agonistas , Péptidos beta-Amiloides/toxicidad , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cumarinas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Membranas Artificiales , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Wortmanina
6.
Oxid Med Cell Longev ; 2021: 3058861, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34812274

RESUMEN

Abnormal accumulations of misfolded Aß and tau proteins are major components of the hallmark plaques and neurofibrillary tangles in the brains of Alzheimer's disease (AD) patients. These abnormal protein deposits cause neurodegeneration through a number of proposed mechanisms, including downregulation of the cAMP-response-element (CRE) binding protein 1 (CREB) signaling pathway. Using CRE-GFP reporter cells, we investigated the effects of three coumarin-chalcone derivatives synthesized in our lab on CREB-mediated gene expression. Aß-GFP- and ΔK280 tauRD-DsRed-expressing SH-SY5Y cells were used to evaluate these agents for possible antiaggregative, antioxidative, and neuroprotective effects. Blood-brain barrier (BBB) penetration was assessed by pharmacokinetic studies in mice. Of the three tested compounds, (E)-3-(3-(4-(dimethylamino)phenyl)acryloyl)-4-hydroxy-2H-chromen-2-one (LM-021) was observed to increase CREB-mediated gene expression through protein kinase A (PKA), Ca2+/calmodulin-dependent protein kinase II (CaMKII), and extracellular signal-regulated kinase (ERK) in CRE-GFP reporter cells. LM-021 exhibited antiaggregative, antioxidative, and neuroprotective effects mediated by the upregulation of CREB phosphorylation and its downstream brain-derived neurotrophic factor and BCL2 apoptosis regulator genes in Aß-GFP- and ΔK280 tauRD-DsRed-expressing SH-SY5Y cells. Blockage of the PKA, CaMKII, or ERK pathway counteracted the beneficial effects of LM-021. LM-021 also exhibited good BBB penetration ability, with brain to plasma ratio of 5.3%, in in vivo pharmacokinetic assessment. Our results indicate that LM-021 works as a CREB enhancer to reduce Aß and tau aggregation and provide neuroprotection. These findings suggest the therapeutic potential of LM-021 in treating AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Chalconas/química , Cumarinas/química , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fármacos Neuroprotectores/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fármacos Neuroprotectores/química , Proteínas tau/genética
7.
Cells ; 10(11)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34831318

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disease presenting with progressive memory and cognitive impairments. One of the pathogenic mechanisms of AD is attributed to the aggregation of misfolded amyloid ß (Aß), which induces neurotoxicity by reducing the expression of brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase B (TRKB) and increasing oxidative stress, caspase-1, and acetylcholinesterase (AChE) activities. Here, we have found the potential of two novel synthetic coumarin derivatives, ZN014 and ZN015, for the inhibition of Aß and neuroprotection in SH-SY5Y neuroblastoma cell models for AD. In SH-SY5Y cells expressing the GFP-tagged Aß-folding reporter, both ZN compounds reduced Aß aggregation, oxidative stress, activities of caspase-1 and AChE, as well as increased neurite outgrowth. By activating TRKB-mediated extracellular signal-regulated kinase (ERK) and AKT serine/threonine kinase 1 (AKT) signaling, these two ZN compounds also upregulated the cAMP-response-element binding protein (CREB) and its downstream BDNF and anti-apoptotic B-cell lymphoma 2 (BCL2). Knockdown of TRKB attenuated the neuroprotective effects of ZN014 and ZN015. A parallel artificial membrane permeability assay showed that ZN014 and ZN015 could be characterized as blood-brain barrier permeable. Our results suggest ZN014 and ZN015 as novel therapeutic candidates for AD and demonstrate that ZN014 and ZN015 reduce Aß neurotoxicity via pleiotropic mechanisms.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Cumarinas/farmacología , Proteínas Fluorescentes Verdes/toxicidad , Fármacos Neuroprotectores/farmacología , Acetilcolinesterasa/metabolismo , Disponibilidad Biológica , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Caspasa 1/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cumarinas/química , Técnicas de Silenciamiento del Gen , Humanos , Proyección Neuronal/efectos de los fármacos , Agregado de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Receptor trkB/metabolismo
8.
Aging (Albany NY) ; 13(11): 15620-15637, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34106880

RESUMEN

Amyloid ß (Aß) plays a major role in the neurodegeneration of Alzheimer's disease (AD). The accumulation of misfolded Aß causes oxidative stress and inflammatory damage leading to apoptotic cell death. Traditional Chinese herbal medicine (CHM) has been widely used in treating neurodegenerative diseases by reducing oxidative stress and neuroinflammation. We examined the neuroprotective effect of formulated CHM Shaoyao Gancao Tang (SG-Tang, made of Paeonia lactiflora and Glycyrrhiza uralensis at 1:1 ratio) in AD cell and mouse models. In Aß-GFP SH-SY5Y cells, SG-Tang reduced Aß aggregation and reactive oxygen species (ROS) production, as well as improved neurite outgrowth. When the Aß-GFP-expressing cells were stimulated with conditioned medium from interferon (IFN)-γ-activated HMC3 microglia, SG-Tang suppressed expressions of inducible nitric oxide synthase (iNOS), NLR family pyrin domain containing 1 (NLRP1) and 3 (NLRP3), tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6, attenuated caspase-1 activity and ROS production, and promoted neurite outgrowth. In streptozocin-induced hyperglycemic APP/PS1/Tau triple transgenic (3×Tg-AD) mice, SG-Tang also reduced expressions of NLRP1, NLRP3, Aß and Tau in hippocampus and cortex, as well as improved working and spatial memories in Y maze and Morris water maze. Collectively, our results demonstrate the potential of SG-Tang in treating AD by moderating neuroinflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Cognición , Medicamentos Herbarios Chinos/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuroprotección , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular , Cognición/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interferón gamma/metabolismo , Memoria/efectos de los fármacos , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Biológicos , Proyección Neuronal/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Agregado de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Aprendizaje Espacial/efectos de los fármacos , Proteínas tau/metabolismo
9.
Aging (Albany NY) ; 12(23): 23619-23646, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33196459

RESUMEN

Polyglutamine (polyQ)-mediated spinocerebellar ataxias (SCA) are caused by mutant genes with expanded CAG repeats encoding polyQ tracts. The misfolding and aggregation of polyQ proteins result in increased reactive oxygen species (ROS) and cellular toxicity. Inflammation is a common manifestation of oxidative stress and inflammatory process further reduces cellular antioxidant capacity. Increase of activated microglia in the pons of SCA type 3 (SCA3) patients suggests the involvement of neuroinflammation in the disease pathogenesis. In this study, we evaluated the anti-inflammatory potentials of indole compound NC009-1, 4-aminophenol-arachidonic acid derivative AM404, quinoline compound VB-037 and chalcone-coumarin derivative LM-031 using human HMC3 microglia and SCA3 ATXN3/Q75-GFP SH-SY5Y cells. The four tested compounds displayed anti-inflammatory activity by suppressing NO, IL-1ß, TNF-α and IL-6 production and CD68 expression of IFN-γ-activated HMC3 microglia. In retinoic acid-differentiated ATXN3/Q75-GFP SH-SY5Y cells inflamed with IFN-γ-primed HMC3 conditioned medium, treatment with the tested compounds mitigated the increased caspase 1 activity and lactate dehydrogenase release, reduced polyQ aggregation and ROS and/or promoted neurite outgrowth. Examination of IL-1ß- and TNF-α-mediated signaling pathways revealed that the tested compounds decreased IκBα/P65, JNK/JUN and/or P38/STAT1 signaling. The study results suggest the potential of NC009-1, AM404, VB-037 and LM-031 in treating SCA3 and probable other polyQ diseases.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Araquidónicos/farmacología , Cumarinas/farmacología , Indoles/farmacología , Enfermedad de Machado-Joseph/tratamiento farmacológico , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Quinolinas/farmacología , Línea Celular Tumoral , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Enfermedad de Machado-Joseph/patología , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
10.
Aging Cell ; 19(7): e13169, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32496635

RESUMEN

Microtubule-associated protein Tau, abundant in the central nervous system (CNS), plays crucial roles in microtubule assembly and stabilization. Abnormal Tau phosphorylation and aggregation are a common pathogenic hallmark in Alzheimer's disease (AD). Hyperphosphorylation of Tau could change its conformation and result in self-aggregation, increased oxidative stress, and neuronal death. In this study, we examined the potential of licochalcone A (a natural chalcone) and five synthetic derivatives (LM compounds) for inhibiting Tau misfolding, scavenging reactive oxygen species (ROS) and providing neuroprotection in human cells expressing proaggregant ΔK280 TauRD -DsRed. All test compounds were soluble up to 100 µM in cell culture media and predicted to be orally bioavailable and CNS-active. Among them, licochalcone A and LM-031 markedly reduced Tau misfolding and associated ROS, promoted neurite outgrowth, and inhibited caspase 3 activity in ΔK280 TauRD -DsRed 293 and SH-SY5Y cells. Mechanistic studies showed that LM-031 upregulates HSPB1 chaperone, NRF2/NQO1/GCLC pathway, and CREB-dependent BDNF/AKT/ERK/BCL2 pathway in ΔK280 TauRD -DsRed SH-SY5Y cells. Decreased neurite outgrowth upon induction of ΔK280 TauRD -DsRed was rescued by LM-031, which was counteracted by knockdown of NRF2 or CREB. LM-031 further rescued the downregulated NRF2 and pCREB, reduced Aß and Tau levels in hippocampus and cortex, and ameliorated cognitive deficits in streptozocin-induced hyperglycemic 3 × Tg-AD mice. Our findings strongly indicate the potential of LM-031 for modifying AD progression by targeting HSPB1 to reduce Tau misfolding and activating NRF2 and CREB pathways to suppress apoptosis and promote neuron survival, thereby offering a new drug development avenue for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Apoptosis , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Estrés Oxidativo , Regulación hacia Arriba
11.
Neurochem Int ; 125: 175-186, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30707915

RESUMEN

The pathogenesis of Alzheimer's disease (AD) is involved in the aggregation of misfolded amyloid ß (Aß), which upregulates the activity of acetylcholinesterase (AChE), increases the production of reactive oxygen species (ROS), enhances neuroinflammation, and eventually leads to neuronal death. Therefore, compounds targeting these mechanisms may be candidates for multitarget drugs in AD treatment. We found that two quinoline derivatives, VB-030 and VB-037, markedly reduced Aß aggregation and ROS levels in the thioflavin T biochemical assay and Tet-On Aß-green fluorescent protein (GFP) 293 AD cell model. These compounds further improved neurite outgrowth, reduced AChE activity and upregulated the molecular chaperone heat shock protein family B [small] member 1 (HSP27), whereas knockdown of HSP27 counteracted the compounds' neuroprotective effects on the Tet-On Aß-GFP SH-SY5Y AD neuronal model. Furthermore, VB-037 attenuated lipopolysaccharide (LPS)/interferon (IFN)-γ-induced activation of BV-2 microglial cells. In addition, VB-037 demonstrated its potential to diminish LPS/IFN-γ-induced upregulation of caspase 1 activity, expression of interleukin (IL)-1ß, and active phosphorylation of mitogen-activated protein kinase 14 (P38), mitogen-activated protein kinase 8 (JNK), and Jun proto-oncogene, AP-1 transcription factor subunit (JUN) signalings, as well as improve cell viability in the Tet-On Aß-GFP SH-SY5Y AD neuronal model. Our findings strongly indicate the potential of VB-037 for modifying AD progression by targeting multiple mechanisms, thereby offering a new drug development avenue for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Mediadores de Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proyección Neuronal/fisiología , Quinolinas/farmacología , Quinolinas/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Línea Celular , Curcumina/farmacología , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proyección Neuronal/efectos de los fármacos , Agregado de Proteínas/efectos de los fármacos , Agregado de Proteínas/fisiología , Proto-Oncogenes Mas , Quinolinas/química , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
J Alzheimers Dis ; 67(2): 737-756, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30689566

RESUMEN

Alzheimer's disease (AD), associated with abnormal accumulation of amyloid-ß (Aß), is the most common cause of dementia among older people. A few studies have identified substantial AD biomarkers in blood but their results were inconsistent. Here we screened gene expression alterations on Aß-GFP SH-SY5Y neuronal model for AD, and evaluated the findings on peripheral leukocytes from 78 patients with AD and 56 healthy controls. The therapeutic responses of identified biomarker candidates were further examined in Aß-GFP SH-SY5Y neuronal and APP/PS1/Tau triple transgenic (3×Tg-AD) mouse models. Downregulation of apolipoprotein E (APOE) and tropomyosin receptor kinase A (TRKA) were detected in Aß-GFP SH-SY5Y cells and validated by peripheral leukocytes from AD patients. Treatment with an in-house indole compound NC009-1 upregulated the expression of APOE and TRKA accompanied with improvement of neurite outgrowth in Aß-GFP SH-SY5Y cells. NC009-1 further rescued the downregulated APOE and TRKA and reduced Aß and tau levels in hippocampus and cortex, and ameliorated cognitive deficits in streptozocin-induced hyperglycemic 3×Tg-AD mice. These results suggest the role of APOE and TRKA as potential peripheral biomarkers in AD, and offer a new drug development target of AD treatment. Further studies of a large series of AD patients will be warranted to verify the findings and confirm the correlation between these markers and therapeutic efficacy.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteínas E/metabolismo , Cognición/efectos de los fármacos , Indoles/farmacología , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptor trkA/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Animales , Conducta Animal , Biomarcadores/análisis , Línea Celular , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos
13.
Am J Chin Med ; : 1-25, 2018 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-30284464

RESUMEN

Amyloid [Formula: see text] (A[Formula: see text]) plays a major role in the pathogenesis of Alzheimer's disease (AD). The accumulation of misfolded A[Formula: see text] causes oxidative and inflammatory damage leading to apoptotic cell death. Chinese herbal medicine (CHM) has been widely used in clinical practice to treat neurodegenerative diseases associated with oxidative stress and neuroinflammation. This study examined the neuroprotection effects of CHM extract Glycyrrhiza inflata (G. inflata) and its active constituents, licochalcone A and liquiritigenin in AD. We examined A[Formula: see text] aggregation inhibition, anti-oxidation and neuroprotection in Tet-On A[Formula: see text]-GFP 293/SH-SY5Y cells and anti-inflammatory potential in lipopolysaccharide (LPS)-stimulated RAW 264.7 and LPS and interferon (IFN)-[Formula: see text] (LPS/IFN-[Formula: see text])-activated BV-2 cells. In addition, we applied conditioned media (CM) of BV-2 cells primed with LPS/IFN-[Formula: see text] to A[Formula: see text]-GFP SH-SY5Y cells to uncover the neuroprotective mechanisms. Our results showed that G. inflata extract and its two constituents displayed potentials of A[Formula: see text] aggregation inhibition and radical-scavenging in biochemical assays, A[Formula: see text] misfolding inhibition and reactive oxygen species (ROS) reduction in A[Formula: see text]-GFP 293 cells, as well as neurite outgrowth promotion, acetylcholinesterase inhibition and SOD2 up-regulation in A[Formula: see text]-GFP SH-SY5Y cells. Meanwhile, both G. inflata extract and its constituents suppressed NO, TNF-[Formula: see text], IL-1[Formula: see text], PGE2 and/or Iba1 productions in inflammation-stimulated RAW 264.7 or BV-2 cells. G. inflata extract and its constituents further protected A[Formula: see text]-GFP SH-SY5Y cells from BV-2 CM-induced cell death by ameliorating reduced BCL2 and attenuating increased IGFBP2, cleaved CASP3, BAD and BAX. Collectively, G. inflata extract, licochalcone A and liquiritigenin display neuroprotection through exerting anti-oxidative and anti-inflammatory activities to suppress neuronal apoptosis.

14.
CNS Neurosci Ther ; 24(12): 1286-1298, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30596401

RESUMEN

BACKGROUND: Aggregation of misfolded amyloid ß (Aß) in senile plaques causes oxidative stress and neuronal death in Alzheimer's disease (AD). Compounds possessing antiaggregation and antioxidant properties are promising candidate compounds for AD treatment. METHODS: We examined the potential of synthetic derivatives of licochalcone A and coumarin for inhibiting Aß aggregation, scavenging reactive oxygen species (ROS), and providing neuroprotection by using biochemical assays and Tet-On Aß-GFP 293/SH-SY5Y cell models for AD. RESULTS: Among test compounds, LM-031, a novel chalcone-coumarin hybrid, inhibited Aß aggregation and scavenged free oxygen radicals. LM-031 markedly reduced Aß misfolding and ROS as well as promoted neurite outgrowth and inhibited acetylcholinesterase in Tet-On Aß-GFP 293/SH-SY5Y cells. Mechanistic studies showed upregulation of the HSPB1 chaperone, NRF2/NQO1/GCLC pathway, and CREB/BDNF/BCL2 pathway. Decreased neurite outgrowth upon the induction of Aß-GFP was rescued by LM-031, which was counteracted by knockdown of HSPB1, NRF2, or CREB. CONCLUSION: Taken together, these findings demonstrate that LM-031 exhibited antiaggregation, antioxidant, and neuroprotective effects against Aß toxicity by enhancing HSPB1 and the NRF2-related antioxidant pathway as well as by activating the CREB-dependent survival and antiapoptosis pathway. These results imply that LM-031 may be a new therapeutic compound for AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antioxidantes/farmacología , Chalcona/farmacología , Cumarinas/farmacología , Agregado de Proteínas/efectos de los fármacos , Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides/genética , Línea Celular Tumoral , Chalcona/química , Cumarinas/química , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Concentración 50 Inhibidora , Neuroblastoma/patología , Proyección Neuronal/efectos de los fármacos , Interferencia de ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transfección
15.
Neuropharmacology ; 101: 309-19, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26362358

RESUMEN

Alzheimer's disease (AD) is the most prevalent form of dementia associated with progressive cognitive decline and memory loss. Extracellular ß-amyloid (Aß) is a major constituent of senile plaques, one of the pathological hallmarks of AD. Aß deposition causes neuronal death via a number of possible mechanisms such as increasing oxidative stress. Therefore therapeutic approaches to identify novel Aß aggregate reducers could be effective for AD treatment. Using a Trx-His-Aß biochemical assay, we screened 11 synthetic indolylquinoline compounds, and found NC009-1, -2, -6 and -7 displaying potential to reduce Aß aggregation. Treating Tet-On Aß-GFP 293 cells with these compounds reduced Aß aggregation and reactive oxygen species. These compounds also promoted neurite outgrowth in Tet-On Aß-GFP SH-SY5Y cells. Furthermore, treatment with above compounds improved neuronal cell viability, neurite outgrowth, and synaptophysin expression level in mouse hippocampal primary culture under oligomeric Aß-induced cytotoxicity. Moreover, the tested NC009-1 significantly ameliorated Aß-induced inhibition of hippocampal long-term potentiation in mouse hippocampal slices. Our results demonstrate how synthetic indolylquinoline compounds are likely to work as chemical chaperones in Aß-aggregation reduction and neuroprotection, providing insight into the possible applications of indolylquinoline compounds in AD treatment.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Quinolinas/farmacología , Péptidos beta-Amiloides/farmacología , Animales , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Especies Reactivas de Oxígeno , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...