Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Curr Med Chem ; 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39229985

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is an irreversible, progressive disorder that profoundly impacts both motor and non-motor functions, thereby significantly diminishing the individual's quality of life. Dihydrosinularin (DHS), a natural bioactive molecule derived from soft corals, exhibits low cytotoxicity and anti-inflammatory properties. However, the therapeutic effects of DHS on neurotoxins and PD are currently unknown. OBJECTIVE: This study investigated whether DHS could mitigate 6-hydroxydopamine (6- OHDA)-induced neurotoxicity and explored the role of neuroprotective PI3K downstream signaling pathways, including that of AKT, ERK, JNK, BCL2, and NFκB, in DHS- mediated neuroprotection. METHOD: We treated the human neuroblastoma cell line, SH-SY5Y, with the neurotoxin 6-OHDA to establish a cellular model of PD. Meanwhile, we assessed the anti-apoptotic and neuroprotective properties of DHS through cell viability, apoptosis, and immunostaining assays. Furthermore, we utilized the PI3K inhibitor LY294002 to validate the therapeutic target of DHS. RESULTS: Based on the physicochemical properties of DHS, it can be inferred that it has promising oral bioavailability and permeability across the blood-brain barrier (BBB). It was demonstrated that DHS upregulates phosphorylated AKT and ERK while downregulating phosphorylated JNK. Consequently, this enhances the expression of BCL2, which exerts a protective effect on neuronal cells by inhibiting caspase activity and preventing cell apoptosis. The inhibition of PI3K significantly reduced the relative protective activity of DHS in 6-OHDA-induced neurotoxicity, suggesting that the neuroprotective effects of DHS are mediated through the activation of PI3K signaling. CONCLUSION: By investigating the mechanisms involved in 6-OHDA-induced neurotoxicity, we provided evidence concerning the therapeutic potential of DHS in neuroprotection. Further research into DHS and its mechanisms of action holds promise for developing novel therapeutic strategies for PD.

2.
Biomolecules ; 13(2)2023 01 23.
Article in English | MEDLINE | ID: mdl-36830589

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Alzheimer Disease/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-akt , Neuroblastoma/metabolism , tau Proteins/metabolism , Receptor, trkB/metabolism , Proto-Oncogene Proteins c-bcl-2
3.
Int J Mol Sci ; 24(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36768965

ABSTRACT

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.


Subject(s)
Parkinson Disease , Mice , Humans , Animals , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/metabolism , Neurotoxins/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , Interleukin-6/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Microglia/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Oxidative Stress , Disease Models, Animal , Mice, Inbred C57BL , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects
4.
Biomol Ther (Seoul) ; 31(3): 285-297, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36646447

ABSTRACT

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.

5.
Int J Mol Sci ; 23(21)2022 Oct 22.
Article in English | MEDLINE | ID: mdl-36361524

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neuroblastoma , Neuroprotective Agents , Tauopathies , Humans , Brain-Derived Neurotrophic Factor/metabolism , tau Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Neuroprotection , Alzheimer Disease/drug therapy , Coumarins/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Neuroblastoma/metabolism , Receptor, trkB/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Caspases
6.
Aging (Albany NY) ; 14(18): 7568-7586, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36170028

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Membrane Glycoproteins/agonists , Neuroblastoma , Neuroprotective Agents , Receptor, trkB/agonists , Amyloid beta-Peptides/toxicity , Brain-Derived Neurotrophic Factor/metabolism , Coumarins , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Membranes, Artificial , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2 , Wortmannin
7.
Oxid Med Cell Longev ; 2021: 3058861, 2021.
Article in English | MEDLINE | ID: mdl-34812274

ABSTRACT

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.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Chalcones/chemistry , Coumarins/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Neuroprotective Agents/pharmacology , tau Proteins/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Humans , Male , Mice , Mice, Inbred ICR , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroprotective Agents/chemistry , tau Proteins/genetics
8.
Cells ; 10(11)2021 11 09.
Article in English | MEDLINE | ID: mdl-34831318

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/toxicity , Coumarins/pharmacology , Green Fluorescent Proteins/toxicity , Neuroprotective Agents/pharmacology , Acetylcholinesterase/metabolism , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caspase 1/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Coumarins/chemistry , Gene Knockdown Techniques , Humans , Neuronal Outgrowth/drug effects , Protein Aggregates , Reactive Oxygen Species/metabolism , Receptor, trkB/metabolism
9.
Aging (Albany NY) ; 13(11): 15620-15637, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34106880

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Apoptosis Regulatory Proteins/metabolism , Cognition , Drugs, Chinese Herbal/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotection , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Cognition/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Interferon-gamma/metabolism , Memory/drug effects , Memory Disorders/complications , Memory Disorders/physiopathology , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Models, Biological , Neuronal Outgrowth/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Protein Aggregates/drug effects , Reactive Oxygen Species/metabolism , Spatial Learning/drug effects , tau Proteins/metabolism
10.
Aging (Albany NY) ; 12(23): 23619-23646, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33196459

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonic Acids/pharmacology , Coumarins/pharmacology , Indoles/pharmacology , Machado-Joseph Disease/drug therapy , Microglia/drug effects , Neurons/drug effects , Quinolines/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Machado-Joseph Disease/metabolism , Machado-Joseph Disease/pathology , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Reactive Oxygen Species/metabolism , Signal Transduction
11.
Aging Cell ; 19(7): e13169, 2020 07.
Article in English | MEDLINE | ID: mdl-32496635

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Animals , Apoptosis , Disease Models, Animal , Humans , Male , Mice , Oxidative Stress , Up-Regulation
12.
Neurochem Int ; 125: 175-186, 2019 05.
Article in English | MEDLINE | ID: mdl-30707915

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , HSP27 Heat-Shock Proteins/metabolism , Inflammation Mediators/metabolism , MAP Kinase Signaling System/physiology , Neuronal Outgrowth/physiology , Quinolines/pharmacology , Quinolines/therapeutic use , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cell Line , Curcumin/pharmacology , Humans , Inflammation Mediators/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Neuronal Outgrowth/drug effects , Protein Aggregates/drug effects , Protein Aggregates/physiology , Proto-Oncogene Mas , Quinolines/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Alzheimers Dis ; 67(2): 737-756, 2019.
Article in English | MEDLINE | ID: mdl-30689566

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Apolipoproteins E/metabolism , Cognition/drug effects , Indoles/pharmacology , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Receptor, trkA/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Animals , Behavior, Animal , Biomarkers/analysis , Cell Line , Female , Gene Expression/drug effects , Humans , Male , Mice , Mice, Transgenic , Neurites/drug effects , Neurons/drug effects
14.
Am J Chin Med ; : 1-25, 2018 Oct 04.
Article in English | MEDLINE | ID: mdl-30284464

ABSTRACT

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.

15.
CNS Neurosci Ther ; 24(12): 1286-1298, 2018 12.
Article in English | MEDLINE | ID: mdl-30596401

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Antioxidants/pharmacology , Chalcone/pharmacology , Coumarins/pharmacology , Protein Aggregates/drug effects , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/genetics , Cell Line, Tumor , Chalcone/chemistry , Coumarins/chemistry , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Inhibitory Concentration 50 , Neuroblastoma/pathology , Neuronal Outgrowth/drug effects , RNA Interference/drug effects , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transfection
16.
Neuropharmacology ; 101: 309-19, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26362358

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Long-Term Potentiation/drug effects , Neuroprotective Agents/pharmacology , Quinolines/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , In Vitro Techniques , Long-Term Potentiation/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Reactive Oxygen Species , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL