Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33805974

ABSTRACT

The semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1) or primary amine oxidase (PrAO), is a deaminating enzyme highly expressed in vessels that generates harmful products as a result of its enzymatic activity. As a multifunctional enzyme, it is also involved in inflammation through its ability to bind and promote the transmigration of circulating leukocytes into inflamed tissues. Inflammation is present in different systemic and cerebral diseases, including stroke and Alzheimer's disease (AD). These pathologies show important affectations on cerebral vessels, together with increased SSAO levels. This review summarizes the main roles of SSAO/VAP-1 in human physiology and pathophysiology and discusses the mechanisms by which it can affect the onset and progression of both stroke and AD. As there is an evident interrelationship between stroke and AD, basically through the vascular system dysfunction, the possibility that SSAO/VAP-1 could be involved in the transition between these two pathologies is suggested. Hence, its inhibition is proposed to be an interesting therapeutical approach to the brain damage induced in these both cerebral pathologies.


Subject(s)
Alzheimer Disease/therapy , Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Cerebrovascular Disorders/therapy , Stroke/therapy , Alzheimer Disease/metabolism , Amines/metabolism , Animals , Cell Adhesion , Cerebral Amyloid Angiopathy/metabolism , Cerebrovascular Disorders/metabolism , Disease Progression , Endothelial Cells/metabolism , Glucose/metabolism , Humans , Inflammation/therapy , Leukocytes/cytology , Mice , Rats , Stroke/metabolism
2.
J Cell Biol ; 219(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32931550

ABSTRACT

Carnitine palmitoyltransferase 1C (CPT1C) is a sensor of malonyl-CoA and is located in the ER of neurons. AMPA receptors (AMPARs) mediate fast excitatory neurotransmission in the brain and play a key role in synaptic plasticity. In the present study, we demonstrate across different metabolic stress conditions that modulate malonyl-CoA levels in cortical neurons that CPT1C regulates the trafficking of the major AMPAR subunit, GluA1, through the phosphatidyl-inositol-4-phosphate (PI(4)P) phosphatase SAC1. In normal conditions, CPT1C down-regulates SAC1 catalytic activity, allowing efficient GluA1 trafficking to the plasma membrane. However, under low malonyl-CoA levels, such as during glucose depletion, CPT1C-dependent inhibition of SAC1 is released, facilitating SAC1's translocation to ER-TGN contact sites to decrease TGN PI(4)P pools and trigger GluA1 retention at the TGN. Results reveal that GluA1 trafficking is regulated by CPT1C sensing of malonyl-CoA and provide the first report of a SAC1 inhibitor. Moreover, they shed light on how nutrients can affect synaptic function and cognition.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Membrane Proteins/genetics , Neurons/metabolism , Receptors, AMPA/genetics , Animals , Brain/metabolism , Glucose/metabolism , Humans , Malonyl Coenzyme A/genetics , Mice , Nutrients/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Transport/genetics , Synaptic Transmission/genetics
3.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32205379

ABSTRACT

The number and function of synaptic AMPA receptors (AMPARs) tightly regulates excitatory synaptic transmission. Current evidence suggests that AMPARs are inserted into the postsynaptic membrane during long-term potentiation (LTP) and are removed from the membrane during long-term depression (LTD). Dephosphorylation of GluA1 at Ser-845 and enhanced endocytosis are critical events in the modulation of LTD. Moreover, changes in scaffold proteins from the postsynaptic density (PSD) could be also related to AMPAR regulation in LTD. In the present study we analyzed the effect of chemical LTD (cLTD) on A-kinase anchoring protein (AKAP)150 and AMPARs levels in mouse-cultured neurons. We show that cLTD induces AKAP150 protein degradation via proteasome, coinciding with GluA1 dephosphorylation at Ser-845 and endocytosis of GluA1-containing AMPARs. Pharmacological inhibition of proteasome activity, but not phosphatase calcineurin (CaN), reverted cLTD-induced AKAP150 protein degradation. Importantly, AKAP150 silencing induced dephosphorylation of GluA1 Ser-845 and GluA1-AMPARs endocytosis while AKAP150 overexpression blocked cLTD-mediated GluA1-AMPARs endocytosis. Our results provide direct evidence that cLTD-induced AKAP150 degradation by the proteasome contributes to synaptic AMPARs endocytosis.


Subject(s)
Long-Term Potentiation , Receptors, AMPA , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Animals , Endocytosis , Mice , Neuronal Plasticity , Synapses/metabolism
4.
Drug Dev Res ; 81(2): 215-231, 2020 04.
Article in English | MEDLINE | ID: mdl-31471933

ABSTRACT

Based on the monoamine oxidase (MAO) inhibition properties of aminoheterocycles with a carbonitrile group we have carried out a systematic exploration to discover new classes of carbonitriles endowed with dual MAO and AChE inhibitory activities, and Aß anti-aggregating properties. Eighty-three nitrile-containing compounds, 13 of which are new, were synthesized and evaluated. in vitro screening revealed that 31, a new compound, presented the best lead for trifunctional inhibition against MAO A (0.34 µM), MAO B (0.26 µM), and AChE (52 µM), while 32 exhibited a lead for selective MAO A (0.12 µM) inhibition coupled to AChE (48 µM) inhibition. Computational analysis revealed that the malononitrile group can find an advantageous position with the aromatic cleft and FAD of MAO A or MAO B. However, the total binding energy can be handicapped by an internal penalty caused by twisting of the ligand molecule and subsequent disruption of the conjugation (32 in MAO B compared to the conjugated 31). Conjugation is also important for AChE as well as the hydrophilic character of malononitrile that allows this group to be in close contact with the aqueous environment as seen for 83. Although the effect of 31 and 32 against Aß1-42 , was very weak, the effect of 63 and 65, and of the new compound 75, indicated that these compounds were able to disaggregate Aß1-42 fibrils. The most effective was 63, a (phenylhydrazinylidene)propanedinitrile derivative that also inhibited MAO A (1.65 µM), making it a potential lead for Alzheimer's disease application.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Nitriles/chemical synthesis , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Computational Biology/methods , Computer Simulation , Humans , Models, Molecular , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Nitriles/chemistry , Nitriles/pharmacology , Structure-Activity Relationship
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2189-2202, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31047972

ABSTRACT

Dysfunctions of the vascular system directly contribute to the onset and progression of Alzheimer's disease (AD). The blood-brain barrier (BBB) shows signs of malfunction at early stages of the disease. When Abeta peptide (Aß) is deposited on brain vessels, it induces vascular degeneration by producing reactive oxygen species and promoting inflammation. These molecular processes are also related to an excessive SSAO/VAP-1 (semicarbazide-sensitive amine oxidase) enzymatic activity, observed in plasma and in cerebrovascular tissue of AD patients. We studied the contribution of vascular SSAO/VAP-1 to the BBB dysfunction in AD using in vitro BBB models. Our results show that SSAO/VAP-1 expression is associated to endothelial activation by altering the release of pro-inflammatory and pro-angiogenic angioneurins, most highly IL-6, IL-8 and VEGF. It is also related to a BBB structure alteration, with a decrease in tight-junction proteins such as zona occludens or claudin-5. Moreover, the BBB function reveals increased permeability and leukocyte adhesion in cells expressing SSAO/VAP-1, as well as an enhancement of the vascular Aß deposition induced by mechanisms both dependent and independent of the enzymatic activity of SSAO/VAP-1. These results reveal an interesting role of vascular SSAO/VAP-1 in BBB dysfunction related to AD progression, opening a new window in the search of alternative therapeutic targets for fighting AD.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Cell Adhesion Molecules/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/cytology , Brain/metabolism , Cell Adhesion , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Mice , Neuroglia/cytology , Neuroglia/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Biochem Pharmacol ; 164: 115-128, 2019 06.
Article in English | MEDLINE | ID: mdl-30954486

ABSTRACT

Uric acid (UA) is a promising protective treatment in ischaemic stroke, but the precise molecular targets underlying its in vivo beneficial actions remain unclear. High concentrations of UA inhibit angiogenesis of cultured endothelial cells via Krüppel-like factor 2 (KLF)-induced downregulation of vascular endothelial growth factor (VEGF), a pro-angiogenic mediator that is able to increase blood-brain barrier (BBB) permeability in acute stroke. Here, we investigated whether UA treatment after ischaemic stroke protects brain endothelial cell functions and modulates the KLF2-VEGF-A axis. Transient intraluminal middle cerebral artery (MCA) occlusion/reperfusion was induced in adult male spontaneously hypertensive (SHR) rats and corresponding normotensive Wistar-Kyoto (WKY) rats. Animals received UA (16 mg/kg) or vehicle (Locke's buffer) i.v. at reperfusion. BBB permeability was evaluated by Evans blue extravasation to the brain and in human cerebral endothelial hCMEC/D3 cells under oxygen-glucose deprivation/re-oxygenation. Circulating VEGF-A levels were measured in rats and acute ischaemic stroke patients from the URICO-ICTUS trial. Angiogenesis progression was assessed in Matrigel-cultured MCA. Worse post-stroke brain damage in SHR than WKY rats was associated with higher hyperaemia at reperfusion, increased Evans blue extravasation, exacerbated MCA angiogenic sprouting, and higher VEGF-A levels. UA treatment reduced infarct volume and Evans blue leakage in both rat strains, improved endothelial cell barrier integrity and KLF2 expression, and lowered VEGF-A levels in SHR rats. Hypertensive stroke patients treated with UA showed lower levels of VEGF-A than patients receiving vehicle. Consistently, UA prevented the enhanced MCA angiogenesis in SHR rats by a mechanism involving KLF2 activation. We conclude that UA treatment after ischaemic stroke upregulates KLF2, reduces VEGF-A signalling, and attenuates brain endothelial cell dysfunctions leading to neuroprotection.


Subject(s)
Blood-Brain Barrier/metabolism , Hypertension/blood , Kruppel-Like Transcription Factors/blood , Stroke/blood , Uric Acid/therapeutic use , Vascular Endothelial Growth Factor A/blood , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biomarkers/blood , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Cell Line , Double-Blind Method , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Hypertension/drug therapy , Hypertension/pathology , Kruppel-Like Transcription Factors/agonists , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/drug therapy , Stroke/pathology , Treatment Outcome , Uric Acid/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Transl Res ; 197: 12-31, 2018 07.
Article in English | MEDLINE | ID: mdl-29653075

ABSTRACT

Inflammation, oxidative stress, and formation of advanced glycated end products (AGEs) and advanced lipoxidation end products (ALEs) are important for atherosclerosis. Vascular adhesion protein-1 (VAP-1) participates in inflammation and has semicarbazide-sensitive amine oxidase (SSAO) activity, which catalyzes oxidative deamination to produce hydrogen peroxide and aldehydes, leading to generation of AGEs and ALEs. However, the effect of VAP-1/SSAO inhibition on atherosclerosis remains controversial, and no studies used coronary angiography to evaluate if plasma VAP-1/SSAO is a biomarker for coronary artery disease (CAD). Here, we examined if plasma VAP-1/SSAO is a biomarker for CAD diagnosed by coronary angiography in humans and investigated the effect of VAP-1/SSAO inhibition by a specific inhibitor PXS-4728A on atherosclerosis in cell and animal models. In the study, VAP-1/SSAO expression was increased in plaques in humans and in apolipoprotein E (ApoE)-deficient mice, and colocalized with vascular endothelial cells and smooth muscle cells (SMCs). Patients with CAD had higher plasma VAP-1/SSAO than those without CAD. Plasma VAP-1/SSAO was positively associated with the extent of CAD. In ApoE-deficient mice, VAP-1/SSAO inhibition reduced atheroma and decreased oxidative stress. VAP-1/SSAO inhibition attenuated the expression of adhesion molecules, chemoattractant proteins, and proinflammatory cytokines in the aorta, and suppressed monocyte adhesion and transmigration across human umbilical vein endothelial cells. Consequently, the expression of markers for macrophage recruitment and activation in plaques was decreased by VAP-1/SSAO inhibition. Besides, VAP-1/SSAO inhibition suppressed proliferation and migration of A7r5 SMC. Our data suggest that plasma VAP-1/SSAO is a novel biomarker for the presence and the extent of CAD in humans. VAP-1/SSAO inhibition by PXS-4728A is a potential treatment for atherosclerosis.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/enzymology , Enzyme Inhibitors/therapeutic use , Semicarbazides/pharmacology , Allylamine/analogs & derivatives , Allylamine/pharmacology , Allylamine/therapeutic use , Animals , Atherosclerosis/blood , Benzamides/pharmacology , Benzamides/therapeutic use , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholesterol , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Oxidative Stress/drug effects , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 542-553, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29175057

ABSTRACT

Beyond cholesterol reduction, statins mediate their beneficial effects on stroke patients through pleiotropic actions. They have shown anti-inflammatory properties by a number of different mechanisms, including the inhibition of NF-κB transcriptional activity and the consequent increase and release of adhesion molecules. We have studied simvastatin's effects on the vascular enzyme semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1), which is involved in stroke-mediated brain injury. SSAO/VAP-1 has leukocyte-binding capacity and mediates the expression of other adhesion proteins through signaling molecules generated by its catalytic activity. Our results indicate that soluble SSAO/VAP-1 is released into the bloodstream after an ischemic stimulus, in parallel with an increase in E-selectin and VCAM-1 and correlating with infarct volume. Simvastatin blocks soluble SSAO/VAP-1 release and prevents E-selectin and VCAM-1 overexpression as well. Simvastatin also effectively blocks SSAO/VAP-1-mediated leukocyte adhesion, although it is not an enzymatic inhibitor of SSAO in vitro. In addition, simvastatin-induced changes in adhesion molecules are greater in human brain endothelial cell cultures expressing SSAO/VAP-1, compared to those not expressing it, indicating some synergic effect with SSAO/VAP-1. We think that part of the beneficial effect of simvastatin in stroke is mediated by the attenuation of the SSAO/VAP-1-dependent inflammatory response.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Brain Ischemia/metabolism , Cell Adhesion Molecules/metabolism , Inflammation/metabolism , Simvastatin/pharmacology , Stroke/metabolism , Animals , Brain/metabolism , Brain Ischemia/drug therapy , Cell Adhesion , Cell Line , Disease Models, Animal , E-Selectin/metabolism , Endothelial Cells , Human Umbilical Vein Endothelial Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation/drug therapy , Male , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Rats , Rats, Wistar , Stroke/pathology , Vascular Cell Adhesion Molecule-1/metabolism
9.
J Pharm Pharmacol ; 69(3): 314-324, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28134992

ABSTRACT

OBJECTIVES: To evaluate the in-vitro and in-vivo effects on monoaminergic neurotransmission of ASS234, a promising multitarget-directed ligand (MTDL), for Alzheimer's disease (AD) therapy. METHODS: In vitro was explored the effect of ASS234 on the monoaminergic metabolism in SH-SY5Y and PC12 cell lines, and remaining activity of both monoamine oxidase (MAO) isoforms was assessed. The corresponding dopamine (DA), homovanillic acid (HVA) and 3,4-dihydroxyphenylacetic acid (DOPAC) and noradrenaline (NA) levels were determined by HPLC-ED. In-vivo experiments were carried out Wistar rats and intracerebral guide cannulas were implanted in the hippocampus and in the prefrontal cortex by sterotaxic coordinates. The day after microdialysis samples were collected and levels of 5-HT, DA and NA were determined by (UHPLC) with electrochemical detector. KEY FINDINGS: ASS234 induced a significant increase in serotonin (5-HT) levels in SH-SY5Y cells. In PC12 cells, ASS234 increased significantly the ratio of dopamine (DA)/(HVA + DOPAC), although no apparent differences in (NA) were observed. By in-vivo microdialysis, ASS234 showed a significant increase in the extracellular levels of 5-HT and NA in hippocampus whereas in the prefrontal cortex, DA and NA also increased significantly. CONCLUSIONS: This study reveals the ability of ASS234 a MTDL compound, to enhance the monoaminergic neurotransmission supporting its potential use in AD therapy.


Subject(s)
Biogenic Monoamines/metabolism , Dopamine/metabolism , Indoles/pharmacology , Monoamine Oxidase/metabolism , Piperidines/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Hippocampus/drug effects , Hippocampus/metabolism , Homovanillic Acid/metabolism , Ligands , Male , Norepinephrine/metabolism , PC12 Cells , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Serotonin/metabolism
10.
Prog Neurobiol ; 151: 4-34, 2017 04.
Article in English | MEDLINE | ID: mdl-26797191

ABSTRACT

Alzheimer's disease is a multifactorial and fatal neurodegenerative disorder characterized by decline of cholinergic function, deregulation of other neurotransmitter systems, ß-amyloid fibril deposition, and ß-amyloid oligomers formation. Based on the involvement of a relevant number of biological systems in Alzheimer's disease progression, multitarget compounds may enable therapeutic efficacy. Accordingly, compounds possessing, besides anticholinergic activity and ß-amyloid aggregation inhibition properties, metal chelating and/or nitric oxide releasing properties with additional antioxidant capacity were developed. Other targets relevant to Alzheimer's disease have also been considered in the last years for producing multitarget compounds such as ß-secretase, monoamino oxidases, serotonin receptors and sigma 1 receptors. The purpose of this review will be to highlight recent reports on the development of multitarget compounds for Alzheimer's disease published within the last years focusing on multifunctional ligands characterized by tacrine-like and donepezil-like structures.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Indans/therapeutic use , Piperidines/therapeutic use , Tacrine/therapeutic use , Animals , Donepezil , Humans , Indans/chemistry , Ligands , Piperidines/chemistry , Tacrine/chemistry
11.
J Psychiatry Neurosci ; 42(1): 59-69, 2017 01.
Article in English | MEDLINE | ID: mdl-27636528

ABSTRACT

BACKGROUND: The heterogeneity of Alzheimer disease requires the development of multitarget drugs for treating the symptoms of the disease and its progression. Both cholinergic and monoamine oxidase dysfunctions are involved in the pathological process. Thus, we hypothesized that the development of therapies focused on these targets might be effective. We have developed and assessed a new product, coded ASS234, a multipotent acetyl and butyrylcholinesterase/monoamine oxidase A-B inhibitor with a potent inhibitory effect on amyloid-ß aggregation as well as antioxidant and antiapoptotic properties. But there is a need to reliably correlate in vitro and in vivo drug release data. METHODS: We examined the effect of ASS234 on cognition in healthy adult C57BL/6J mice in a model of scopolamine-induced cognitive impairment that often accompanies normal and pathological aging. Also, in a characterized transgenic APPswe/PS1ΔE9 mouse model of Alzheimer disease, we examined the effects of short-term ASS234 treatment on plaque deposition and gliosis using immunohistochemistry. Toxicology of ASS234 was assessed using a quantitative high-throughput in vitro cytotoxicity screening assay following the MTT assay method in HepG2 liver cells. RESULTS: In vivo, ASS234 significantly decreased scopolamine-induced learning deficits in C57BL/6J mice. Also, reduction of amyloid plaque burden and gliosis in the cortex and hippocampus was assessed. In vitro, ASS234 exhibited lesser toxicity than donepezil and tacrine. LIMITATIONS: The study was conducted in male mice only. Although the Alzheimer disease model does not recapitulate all features of the human disease, it exhibits progressive monoaminergic neurodegeneration. CONCLUSION: ASS234 is a promising alternative drug of choice to treat the cognitive decline and neurodegeneration underlying Alzheimer disease.


Subject(s)
Alzheimer Disease/drug therapy , Cerebral Cortex/drug effects , Hippocampus/drug effects , Indoles/administration & dosage , Learning/drug effects , Nootropic Agents/administration & dosage , Piperidines/administration & dosage , Alzheimer Disease/pathology , Animals , Cell Survival/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Donepezil , Gliosis/drug therapy , Gliosis/pathology , Hep G2 Cells , Hippocampus/metabolism , Humans , Indans/toxicity , Indoles/chemistry , Indoles/toxicity , Male , Mice, Inbred C57BL , Nootropic Agents/chemistry , Nootropic Agents/toxicity , Piperidines/chemistry , Piperidines/toxicity , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Proof of Concept Study , Recognition, Psychology/drug effects , Scopolamine , Tacrine/toxicity
12.
Front Neurosci ; 10: 541, 2016.
Article in English | MEDLINE | ID: mdl-27932945

ABSTRACT

The monoaminergic systems are the target of several drugs for the treatment of mood, motor and cognitive disorders as well as neurological conditions. In most cases, advances have occurred through serendipity, except for Parkinson's disease where the pathophysiology led almost immediately to the introduction of dopamine restoring agents. Extensive neuropharmacological studies first showed that the primary target of antipsychotics, antidepressants, and anxiolytic drugs were specific components of the monoaminergic systems. Later, some dramatic side effects associated with older medicines were shown to disappear with new chemical compounds targeting the origin of the therapeutic benefit more specifically. The increased knowledge regarding the function and interaction of the monoaminergic systems in the brain resulting from in vivo neurochemical and neurophysiological studies indicated new monoaminergic targets that could achieve the efficacy of the older medicines with fewer side-effects. Yet, this accumulated knowledge regarding monoamines did not produce valuable strategies for diseases where no monoaminergic drug has been shown to be effective. Here, we emphasize the new therapeutic and monoaminergic-based strategies for the treatment of psychiatric diseases. We will consider three main groups of diseases, based on the evidence of monoamines involvement (schizophrenia, depression, obesity), the identification of monoamines in the diseases processes (Parkinson's disease, addiction) and the prospect of the involvement of monoaminergic mechanisms (epilepsy, Alzheimer's disease, stroke). In most cases, the clinically available monoaminergic drugs induce widespread modifications of amine tone or excitability through neurobiological networks and exemplify the overlap between therapeutic approaches to psychiatric and neurological conditions. More recent developments that have resulted in improved drug specificity and responses will be discussed in this review.

13.
Front Neurosci ; 10: 294, 2016.
Article in English | MEDLINE | ID: mdl-27445665

ABSTRACT

HIGHLIGHTS: ASS2324 is a hybrid compound resulting from the juxtaposition of donepezil and the propargylamine PF9601N ASS2324 is a multi-target directed propargylamine able to bind to all the AChE/BuChE and MAO A/B enzymesASS2324 shows antioxidant, neuroprotective and suitable permeability propertiesASS2324 restores the scopolamine-induced cognitive impairment to the same extent as donepezil, and is less toxicASS2324 prevents ß-amyloid induced aggregation in the cortex of double transgenic miceASS2324 is the most advanced anti-Alzheimer agent for pre-clinical studies that we have identified in our laboratories The complex nature of Alzheimer's disease (AD) has prompted the design of Multi-Target-Directed Ligands (MTDL) able to bind to diverse biochemical targets involved in the progress and development of the disease. In this context, we have designed a number of MTD propargylamines (MTDP) showing antioxidant, anti-beta-amyloid, anti-inflammatory, as well as cholinesterase and monoamine oxidase (MAO) inhibition capacities. Here, we describe these properties in the MTDL ASS234, our lead-compound ready to enter in pre-clinical studies for AD, as a new multipotent, permeable cholinesterase/monoamine oxidase inhibitor, able to inhibit Aß-aggregation, and possessing antioxidant and neuroprotective properties.

14.
Bioorg Med Chem ; 24(20): 4835-4854, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27396685

ABSTRACT

Different azides and alkynes have been coupled via Cu-catalyzed 1,3-dipolar Huisgen cycloaddition to afford a novel family of N1- and C5-substituted 1,2,3-triazole derivatives that feature the propargylamine group typical of irreversible MAO-B inhibitors at the C4-side chain of the triazole ring. All the synthesized compounds were evaluated against human MAO-A and MAO-B. Structure-activity relationships and molecular modeling were utilized to gain insight into the structural and chemical features that enhance the binding affinity and selectivity between the two enzyme isoforms. Several lead compounds, in terms of potency (submicromolar to low micromolar range), MAO-B selective recognition, and brain permeability, were identified. One of these leads (MAO-B IC50 of 3.54µM, selectivity MAO-A/MAO-B index of 27.7) was further subjected to reversibility and time-dependence inhibition studies, which disclosed a slow and irreversible inhibition of human MAO-B. Overall, the results support the suitability of the 4-triazolylalkyl propargylamine scaffold for exploring the design of multipotent anti-Alzheimer compounds endowed with irreversible MAO-B inhibitory activity.


Subject(s)
Drug Design , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pargyline/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Monoamine Oxidase Inhibitors/chemistry , Pargyline/analogs & derivatives , Pargyline/chemical synthesis , Pargyline/chemistry , Structure-Activity Relationship
15.
Front Neurosci ; 10: 205, 2016.
Article in English | MEDLINE | ID: mdl-27252617

ABSTRACT

HIGHLIGHTS ASS234 is a MTDL compound containing a moiety from Donepezil and the propargyl group from the PF 9601N, a potent and selective MAO B inhibitor. This compound is the most advanced anti-Alzheimer agent for preclinical studies identified in our laboratory.Derived from ASS234 both multipotent donepezil-indolyl (MTDL-1) and donepezil-pyridyl hybrids (MTDL-2) were designed and evaluated as inhibitors of AChE/BuChE and both MAO isoforms. MTDL-2 showed more high affinity toward the four enzymes than MTDL-1.MTDL-3 and MTDL-4, were designed containing the N-benzylpiperidinium moiety from Donepezil, a metal- chelating 8-hydroxyquinoline group and linked to a N-propargyl core and they were pharmacologically evaluated.The presence of the cyano group in MTDL-3, enhanced binding to AChE, BuChE and MAO A. It showed antioxidant behavior and it was able to strongly complex Cu(II), Zn(II) and Fe(III).MTDL-4 showed higher affinity toward AChE, BuChE.MTDL-3 exhibited good brain penetration capacity (ADMET) and less toxicity than Donepezil. Memory deficits in scopolamine-lesioned animals were restored by MTDL-3.MTDL-3 particularly emerged as a ligand showing remarkable potential benefits for its use in AD therapy. Alzheimer's disease (AD), the most common form of adult onset dementia, is an age-related neurodegenerative disorder characterized by progressive memory loss, decline in language skills, and other cognitive impairments. Although its etiology is not completely known, several factors including deficits of acetylcholine, ß-amyloid deposits, τ-protein phosphorylation, oxidative stress, and neuroinflammation are considered to play significant roles in the pathophysiology of this disease. For a long time, AD patients have been treated with acetylcholinesterase inhibitors such as donepezil (Aricept®) but with limited therapeutic success. This might be due to the complex multifactorial nature of AD, a fact that has prompted the design of new Multi-Target-Directed Ligands (MTDL) based on the "one molecule, multiple targets" paradigm. Thus, in this context, different series of novel multifunctional molecules with antioxidant, anti-amyloid, anti-inflammatory, and metal-chelating properties able to interact with multiple enzymes of therapeutic interest in AD pathology including acetylcholinesterase, butyrylcholinesterase, and monoamine oxidases A and B have been designed and assessed biologically. This review describes the multiple targets, the design rationale and an in-house MTDL library, bearing the N-benzylpiperidine motif present in donepezil, linked to different heterocyclic ring systems (indole, pyridine, or 8-hydroxyquinoline) with special emphasis on compound ASS234, an N-propargylindole derivative. The description of the in vitro biological properties of the compounds and discussion of the corresponding structure-activity-relationships allows us to highlight new issues for the identification of more efficient MTDL for use in AD therapy.

16.
Eur J Med Chem ; 121: 864-879, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-26471320

ABSTRACT

Currently available drugs against Alzheimer's disease (AD) are only able to ameliorate the disease symptoms resulting in a moderate improvement in memory and cognitive function without any efficacy in preventing and inhibiting the progression of the pathology. In an effort to obtain disease-modifying anti-Alzheimer's drugs (DMAADs) following the multifactorial nature of AD, we have recently developed multifunctional compounds. We herein describe the design, synthesis, molecular modeling and biological evaluation of a new series of donepezil-related compounds possessing metal chelating properties, and being capable of targeting different enzymatic systems related to AD (cholinesterases, ChEs, and monoamine oxidase A, MAO-A). Among this set of analogues compound 5f showed excellent ChEs inhibition potency and a selective MAO-A inhibition (vs MAO-B) coupled to strong complexing properties for zinc and copper ions, both known to be involved in the progression of AD. Moreover, 5f exhibited moderate antioxidant properties as found by in vitro assessment. This compound represents a novel donepezil-hydroxyquinoline hybrid with DMAAD profile paving the way to the development of a novel class of drugs potentially able to treat AD.


Subject(s)
Acetylcholinesterase/metabolism , Drug Design , Indans/chemical synthesis , Indans/pharmacology , Molecular Docking Simulation , Monoamine Oxidase/metabolism , Piperidines/chemical synthesis , Piperidines/pharmacology , Acetylcholinesterase/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Antioxidants/metabolism , Antioxidants/pharmacology , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Chelating Agents/metabolism , Chelating Agents/pharmacology , Chemistry Techniques, Synthetic , Donepezil , Humans , Indans/chemistry , Indans/metabolism , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Piperidines/chemistry , Piperidines/metabolism , Protein Conformation , Structure-Activity Relationship
17.
Food Funct ; 6(4): 1251-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25756794

ABSTRACT

The possible modulatory effect of the functional LMN diet, rich in theobromine, polyphenols and polyunsaturated fatty acids, on the catecholaminergic and cholinergic neurotransmission, affecting cognition decline during aging has been studied. 129S1/SvlmJ mice were fed for 10, 20, 30 and 40 days with either LMN or control diets. The enzymes involved in catecholaminergic and cholinergic metabolism were determined by both immunohistological and western blot analyses. Noradrenalin, dopamine and other metabolites were quantified by HPLC analysis. Theobromine, present in cocoa, the main LMN diet component, was analysed in parallel using SH-SY5Y and PC12 cell lines. An enhanced modulatory effect on both cholinergic and catecholaminergic transmissions was observed on 20 day fed mice. Similar effect was observed with theobromine, besides its antioxidant capacity inducing SOD-1 and GPx expression. The enhancing effect of the LMN diet and theobromine on the levels of acetylcholine-related enzymes, dopamine and specially noradrenalin confirms the beneficial role of this diet on the "cognitive reserve" and hence a possible reducing effect on cognitive decline underlying aging and Alzheimer's disease.


Subject(s)
Aging/drug effects , Cholinergic Neurons/drug effects , Fatty Acids, Unsaturated/administration & dosage , Hippocampus/drug effects , Polyphenols/administration & dosage , Theobromine/administration & dosage , Acetylcholinesterase/metabolism , Alzheimer Disease/prevention & control , Animals , Cacao/chemistry , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/metabolism , Chromatography, High Pressure Liquid , Cognition/drug effects , Diet , Dopamine/metabolism , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Mice , Mice, 129 Strain , Norepinephrine/metabolism , PC12 Cells , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
18.
Neurobiol Aging ; 36(2): 762-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25457560

ABSTRACT

An association between semicarbazide-sensitive amine oxidase (SSAO) and cerebral amyloid angiopathy (CAA) related to Alzheimer's disease (AD) has been largely postulated. Increased SSAO activity and expression have been detected in cerebrovascular tissue and plasma of AD patients, colocalizing with cerebrovascular amyloid-beta (Aß) deposits. As an enzyme, SSAO metabolizes primary amines generating hydrogen peroxide, ammonia, and aldehydes. The ability of these products to generate oxidative stress, to enhance the advanced glycation end-product generation, to promote the Aß aggregation in vitro, and to induce apoptosis supports its role in CAA-related vascular pathology. However, whether the SSAO increase constitutes a cause or it is a consequence of the pathologic process has not been elucidated so far. To set up the nature of this relationship, vascular cell models expressing SSAO were treated with different Aß forms, simulating the CAA conditions in vitro. It was found that the presence of the vasculotropic Dutch-mutated Aß1-40 increases (Aß1-40 D) the SSAO-dependent toxicity, which is accompanied by an increase of SSAO protein availability in endothelial cell membranes. In addition, SSAO enhances Aß1-40 D and Aß1-42 deposition on vascular cells by both activity-dependent and -independent mechanisms. Thus, we provide evidences indicating that Aß itself could be one of the factors inducing SSAO increase in AD, enhancing its toxic effect, and inducing the vascular dysfunction and, in turn, that SSAO stimulates Aß deposition on the vascular walls, thereby contributing to the CAA-AD progression. Therefore, molecules inhibiting SSAO could provide an alternative treatment for preventing/delaying the progress of CAA-AD-associated vasculopathy.


Subject(s)
Alzheimer Disease/genetics , Amine Oxidase (Copper-Containing)/physiology , Amyloid beta-Peptides/metabolism , Cell Adhesion Molecules/physiology , Cerebral Amyloid Angiopathy/genetics , Endothelial Cells/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amine Oxidase (Copper-Containing)/genetics , Amine Oxidase (Copper-Containing)/metabolism , Apoptosis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/therapy , Endothelial Cells/metabolism , Gene Expression , Glycation End Products, Advanced/metabolism , Humans , Molecular Targeted Therapy , Oxidative Stress
19.
Drug Des Devel Ther ; 8: 1893-910, 2014.
Article in English | MEDLINE | ID: mdl-25378907

ABSTRACT

The design, synthesis, and biochemical evaluation of donepezil-pyridyl hybrids (DPHs) as multipotent cholinesterase (ChE) and monoamine oxidase (MAO) inhibitors for the potential treatment of Alzheimer's disease (AD) is reported. The 3D-quantitative structure-activity relationship study was used to define 3D-pharmacophores for inhibition of MAO A/B, acetylcholinesterase (AChE), and butyrylcholinesterase (BuChE) enzymes and to design DPHs as novel multi-target drug candidates with potential impact in the therapy of AD. DPH14 (Electrophorus electricus AChE [EeAChE]: half maximal inhibitory concentration [IC50] =1.1±0.3 nM; equine butyrylcholinesterase [eqBuChE]: IC50 =600±80 nM) was 318-fold more potent for the inhibition of AChE, and 1.3-fold less potent for the inhibition of BuChE than the reference compound ASS234. DPH14 is a potent human recombinant BuChE (hBuChE) inhibitor, in the same range as DPH12 or DPH16, but 13.1-fold less potent than DPH15 for the inhibition of human recombinant AChE (hAChE). Compared with donepezil, DPH14 is almost equipotent for the inhibition of hAChE, and 8.8-fold more potent for hBuChE. Concerning human monoamine oxidase (hMAO) A inhibition, only DPH9 and 5 proved active, compound DPH9 being the most potent (IC50 [MAO A] =5,700±2,100 nM). For hMAO B, only DPHs 13 and 14 were moderate inhibitors, and compound DPH14 was the most potent (IC50 [MAO B] =3,950±940 nM). Molecular modeling of inhibitor DPH14 within EeAChE showed a binding mode with an extended conformation, interacting simultaneously with both catalytic and peripheral sites of EeAChE thanks to a linker of appropriate length. Absortion, distribution, metabolism, excretion and toxicity analysis showed that structures lacking phenyl-substituent show better druglikeness profiles; in particular, DPHs13-15 showed the most suitable absortion, distribution, metabolism, excretion and toxicity properties. Novel donepezil-pyridyl hybrid DPH14 is a potent, moderately selective hAChE and selective irreversible hMAO B inhibitor which might be considered as a promising compound for further development for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Drug Design , Indans/therapeutic use , Monoamine Oxidase Inhibitors/therapeutic use , Piperidines/therapeutic use , Pyridines/therapeutic use , Quantitative Structure-Activity Relationship , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterases/metabolism , Donepezil , Humans , Indans/chemistry , Indans/metabolism , Models, Molecular , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Piperidines/chemistry , Piperidines/metabolism , Pyridines/chemistry , Pyridines/metabolism , Recombinant Proteins/metabolism
20.
Eur J Med Chem ; 80: 543-61, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24813882

ABSTRACT

The synthesis, biochemical evaluation, ADMET, toxicity and molecular modeling of novel multi-target-directed Donepezil + Propargylamine + 8-Hydroxyquinoline (DPH) hybrids 1-7 for the potential prevention and treatment of Alzheimer's disease is described. The most interesting derivative was racemic α-aminotrile4-(1-benzylpiperidin-4-yl)-2-(((8-hydroxyquinolin-5-yl)methyl)(prop-2-yn-1-yl)amino) butanenitrile (DPH6) [MAO A (IC50 = 6.2 ± 0.7 µM; MAO B (IC50 = 10.2 ± 0.9 µM); AChE (IC50 = 1.8 ± 0.1 µM); BuChE (IC50 = 1.6 ± 0.25 µM)], an irreversible MAO A/B inhibitor and mixed-type AChE inhibitor with metal-chelating properties. According to docking studies, both DPH6 enantiomers interact simultaneously with the catalytic and peripheral site of EeAChE through a linker of appropriate length, supporting the observed mixed-type AChE inhibition. Both enantiomers exhibited a relatively similar position of both hydroxyquinoline and benzyl moieties with the rest of the molecule easily accommodated in the relatively large cavity of MAO A. For MAO B, the quinoline system was hosted at the cavity entrance whereas for MAO A this system occupied the substrate cavity. In this disposition the quinoline moiety interacted directly with the FAD aromatic ring. Very similar binding affinity values were also observed for both enantiomers with ChE and MAO enzymes. DPH derivatives exhibited moderate to good ADMET properties and brain penetration capacity for CNS activity. DPH6 was less toxic than donepezil at high concentrations; while at low concentrations both displayed a similar cell viability profile. Finally, in a passive avoidance task, the antiamnesic effect of DPH6 was tested on mice with experimentally induced amnesia. DPH6 was capable to significantly decrease scopolamine-induced learning deficits in healthy adult mice.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , Chelating Agents/metabolism , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Chelating Agents/toxicity , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/therapeutic use , Cholinesterase Inhibitors/toxicity , Donepezil , Hep G2 Cells , Humans , Hydroxyquinolines/chemistry , Indans/chemistry , Male , Memory/drug effects , Molecular Docking Simulation , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/therapeutic use , Monoamine Oxidase Inhibitors/toxicity , Pargyline/analogs & derivatives , Pargyline/chemistry , Piperidines/chemistry , Propylamines/chemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...