Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 140, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38291108

ABSTRACT

Plasma-derived therapeutic proteins are produced through an industrial fractionation process where proteins are purified from individual intermediates, some of which remain unused and are discarded. Relatively few plasma-derived proteins are exploited clinically, with most of available plasma being directed towards the manufacture of immunoglobulin and albumin. Although the plasma proteome provides opportunities to develop novel protein replacement therapies, particularly for rare diseases, the high cost of plasma together with small patient populations impact negatively on the development of plasma-derived orphan drugs. Enabling therapeutics development from unused plasma fractionation intermediates would therefore constitute a substantial innovation. To this objective, we characterized the proteome of unused plasma fractionation intermediates and prioritized proteins for their potential as new candidate therapies for human disease. We selected ceruloplasmin, a plasma ferroxidase, as a potential therapy for aceruloplasminemia, an adult-onset ultra-rare neurological disease caused by iron accumulation as a result of ceruloplasmin mutations. Intraperitoneally administered ceruloplasmin, purified from an unused plasma fractionation intermediate, was able to prevent neurological, hepatic and hematological phenotypes in ceruloplasmin-deficient mice. These data demonstrate the feasibility of transforming industrial waste plasma fraction into a raw material for manufacturing of new candidate proteins for replacement therapies, optimizing plasma use and reducing waste generation.


Subject(s)
Ceruloplasmin , Iron Metabolism Disorders , Neurodegenerative Diseases , Proteome , Adult , Humans , Animals , Mice , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Proteome/metabolism , Rare Diseases , Industrial Waste
2.
Int J Mol Sci ; 22(3)2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33494384

ABSTRACT

The onset of chemotherapy-induced peripheral neurotoxicity (CIPN) is a leading cause of the dose reduction or discontinuation of cancer treatment due to sensory symptoms. Paclitaxel (PTX) can cause painful peripheral neuropathy, with a negative impact on cancer survivors' quality of life. While recent studies have shown that neuroinflammation is involved in PTX-induced peripheral neurotoxicity (PIPN), the pathophysiology of this disabling side effect remains largely unclear and no effective therapies are available. Therefore, here we investigated the effects of human intravenous immunoglobulin (IVIg) on a PIPN rat model. PTX-treated rats showed mechanical allodynia and neurophysiological alterations consistent with a severe sensory axonal polyneuropathy. In addition, morphological evaluation showed a reduction of intra-epidermal nerve fiber (IENF) density and evidenced axonopathy with macrophage infiltration, which was more prominent in the distal segment of caudal nerves. Three weeks after the last PTX injection, mechanical allodynia was still present in PTX-treated rats, while the full recovery in the group of animals co-treated with IVIg was observed. At the pathological level, this behavioral result was paralleled by prevention of the reduction in IENF density induced by PTX in IVIg co-treated rats. These results suggest that the immunomodulating effect of IVIg co-treatment can alleviate PIPN neurotoxic manifestations, probably through a partial reduction of neuroinflammation.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Immunoglobulins, Intravenous/administration & dosage , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/etiology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Axons/drug effects , Axons/metabolism , Axons/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility , Humans , Hyperalgesia/diagnosis , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Neurotoxicity Syndromes/diagnosis , Paclitaxel/therapeutic use , Peripheral Nervous System Diseases/diagnosis , Rats , Treatment Outcome
3.
J Neuroinflammation ; 15(1): 232, 2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30131066

ABSTRACT

BACKGROUND: Chemotherapy-induced peripheral neurotoxicity (CIPN) is a severe adverse effect in patients receiving antitumor agents, and no effective treatment is available. Although the mechanisms responsible for the development of CIPN are poorly understood, recent findings make neuroinflammation an attractive target to be investigated, particularly when neuropathic pain is a prominent feature such as after bortezomib administration. The aim of our study was to evaluate the effect of intravenous immunoglobulins (IVIg) delivery in chronic CIPN. The related neuro-immune aspects were investigated in a well-characterized rat model of bortezomib-induced peripheral neurotoxicity (BIPN). METHODS: After determination of a suitable schedule based on a preliminary pharmacokinetic pilot study, female Wistar rats were treated with IVIg 1 g/kg every 2 weeks. IVIg treatment was started at the beginning of bortezomib administration ("preventive" schedule), or once BIPN was already ensued after 4 weeks of treatment ("therapeutic" schedule). Neurophysiological and behavioral studies were performed to assess the extent of painful peripheral neurotoxicity induced by bortezomib, and these functional assessments were completed by pathologic examination of peripheral nerves and intraepidermal nerve fiber quantification (IENF). The role of the innate immune response in BIPN was investigated by immunochemistry characterization of macrophage infiltration in peripheral nerves. RESULTS: Both schedules of IVIg administration were able to significantly reduce bortezomib-induced heat and mechanical allodynia. Although these changes were not evidenced at the neurophysiological examination of peripheral nerves, they behavioral effects were paralleled in the animals treated with the preventive schedule by reduced axonopathy in peripheral nerves and significant protection from loss of IENF. Moreover, IVIg administration was very effective in reducing infiltration in peripheral nerves of macrophages with the M1, pro-inflammatory phenotype. CONCLUSION: Our results suggest a prominent role of neuroinflammation in BIPN and that IVIg might be considered as a possible safe and effective therapeutic option preventing M1 macrophage infiltration. However, since neuropathic pain is frequent also in other CIPN types, it also indicates the need for further investigation in other forms of CIPN.


Subject(s)
Immunoglobulins/therapeutic use , Immunologic Factors/therapeutic use , Macrophages/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Peripheral Nerves/pathology , Animals , Antineoplastic Agents/toxicity , Body Weight/drug effects , Bortezomib/toxicity , Cytokines/metabolism , Disease Models, Animal , Hot Temperature/adverse effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Macrophages/pathology , Nerve Fibers/drug effects , Nerve Fibers/pathology , Neural Conduction/drug effects , Neurotoxicity Syndromes/etiology , Neutrophil Infiltration , Physical Stimulation/adverse effects , Rats , Sensory Thresholds/drug effects , Skin/pathology
4.
Eur J Pharm Sci ; 86: 136-42, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-26987608

ABSTRACT

Preclinical imaging modalities represent an essential tool to develop a modern and translational biomedical research. To date, Optical Imaging (OI) and Magnetic Resonance Imaging (MRI) are used principally in separate studies for molecular imaging studies. We decided to combine OI and MRI together through the development of a lentiviral vector to monitor the Wnt pathway response to Lithium Chloride (LiCl) treatment. The construct was stably infected in glioblastoma cells and, after intracranial transplantation in mice, serial MRI and OI imaging sessions were performed to detect human ferritin heavy chain protein (hFTH) and firefly luciferase enzyme (FLuc) respectively. The system allowed also ex vivo analysis using a constitutive fluorescence protein expression. In mice, LiCl administration has shown significantly increment of luminescence signal and a lower signal of T2 values (P<0.05), recorded noninvasively with OI and a 7 Tesla MRI scanner. This study indicates that OI and MRI can be performed in a single in vivo experiment, providing an in vivo proof-of-concept for drug discovery projects in preclinical phase.


Subject(s)
Genes, Reporter/genetics , Molecular Imaging , Animals , Apoferritins/genetics , Apoferritins/metabolism , Brain/metabolism , Cell Line, Tumor , Female , Gene Expression , Humans , Lithium Chloride/pharmacology , Luciferases, Firefly/genetics , Magnetic Resonance Imaging , Mice, Nude , Optical Imaging , Wnt Signaling Pathway
5.
Neurobiol Dis ; 86: 41-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26626080

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by dyskinesia, cognitive impairment and emotional disturbances, presenting progressive neurodegeneration in the striatum and intracellular mutant Huntingtin (mHTT) aggregates in various areas of the brain. Recombinant Adeno Associated Viral (rAAV) vectors have been successfully used to transfer foreign genes to the brain of adult animals. In the present study we report a novel in vivo rat HD model obtained by stereotaxic injection of rAAV serotype2/9 containing Exon1-Q138 mHTT (Q138) and Exon1-Q17 wild type HTT (Q17; control), respectively in the right and in the left striatum, and expressed as C-terminal GFP fusions to facilitate detection of infected cells and aggregate production. Immunohistochemical analysis of brain slices from animals sacrificed twenty-one days after viral infection showed that Q138 injection resulted in robust formation of GFP-positive aggregates in the striatum, increased GFAP and microglial activation and neurodegeneration, with little evidence of any of these events in contralateral tissue infected with wild type (Q17) expressing construct. Differences in the relative metabolite concentrations (N-Acetyl Aspartate/Creatine and Myo-Inositol/Creatine) were observed by H1 MR Spectroscopy. By quantitative RT-PCR we also demonstrated that mHTT induced changes in the expression of genes previously shown to be altered in other rodent HD models. Importantly, administration of reference compounds previously shown to ameliorate the aggregation and neurodegeneration phenotypes in preclinical HD models was demonstrated to revert the mutant HTT-dependent effects in our model. In conclusion, the AAV2/9-Q138/Q17 exon 1 HTT stereotaxic injection represents a useful first-line in vivo preclinical model for studying the biology of mutant HTT exon 1 in the striatum and to provide early evidence of efficacy of therapeutic approaches.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/virology , Dependovirus/genetics , Disease Models, Animal , Drug Discovery/methods , Genetic Vectors/administration & dosage , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Corpus Striatum/pathology , Encephalitis/metabolism , Encephalitis/virology , Exons , Female , Green Fluorescent Proteins/metabolism , Huntingtin Protein , Huntington Disease/metabolism , Neuroglia/metabolism , Neurons/pathology , Neurons/virology , Rats , Rats, Wistar , Recombinant Proteins/metabolism
6.
Nat Chem Biol ; 11(5): 347-354, 2015 May.
Article in English | MEDLINE | ID: mdl-25848931

ABSTRACT

Huntington's disease (HD) is a currently incurable neurodegenerative condition caused by an abnormally expanded polyglutamine tract in huntingtin (HTT). We identified new modifiers of mutant HTT toxicity by performing a large-scale 'druggable genome' siRNA screen in human cultured cells, followed by hit validation in Drosophila. We focused on glutaminyl cyclase (QPCT), which had one of the strongest effects on mutant HTT-induced toxicity and aggregation in the cell-based siRNA screen and also rescued these phenotypes in Drosophila. We found that QPCT inhibition induced the levels of the molecular chaperone αB-crystallin and reduced the aggregation of diverse proteins. We generated new QPCT inhibitors using in silico methods followed by in vitro screening, which rescued the HD-related phenotypes in cell, Drosophila and zebrafish HD models. Our data reveal a new HD druggable target affecting mutant HTT aggregation and provide proof of principle for a discovery pipeline from druggable genome screen to drug development.


Subject(s)
Aminoacyltransferases/drug effects , Aminoacyltransferases/genetics , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA, Small Interfering , Aminoacyltransferases/antagonists & inhibitors , Animals , Cells, Cultured , Computational Biology , Drosophila , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Zebrafish , alpha-Crystallin B Chain/metabolism
7.
Hum Mol Genet ; 23(11): 2995-3007, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24436303

ABSTRACT

Protein acetylation, which is central to transcriptional control as well as other cellular processes, is disrupted in Huntington's disease (HD). Treatments that restore global acetylation levels, such as inhibiting histone deacetylases (HDACs), are effective in suppressing HD pathology in model organisms. However, agents that selectively target the disease-relevant HDACs have not been available. SirT1 (Sir2 in Drosophila melanogaster) deacetylates histones and other proteins including transcription factors. Genetically reducing, but not eliminating, Sir2 has been shown to suppress HD pathology in model organisms. To date, small molecule inhibitors of sirtuins have exhibited low potency and unattractive pharmacological and biopharmaceutical properties. Here, we show that highly selective pharmacological inhibition of Drosophila Sir2 and mammalian SirT1 using the novel inhibitor selisistat (selisistat; 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide) can suppress HD pathology caused by mutant huntingtin exon 1 fragments in Drosophila, mammalian cells and mice. We have validated Sir2 as the in vivo target of selisistat by showing that genetic elimination of Sir2 eradicates the effect of this inhibitor in Drosophila. The specificity of selisistat is shown by its effect on recombinant sirtuins in mammalian cells. Reduction of HD pathology by selisistat in Drosophila, mammalian cells and mouse models of HD suggests that this inhibitor has potential as an effective therapeutic treatment for human disease and may also serve as a tool to better understand the downstream pathways of SirT1/Sir2 that may be critical for HD.


Subject(s)
Carbazoles/administration & dosage , Drosophila Proteins/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Huntington Disease/drug therapy , Huntington Disease/enzymology , Sirtuin 1/antagonists & inhibitors , Sirtuins/antagonists & inhibitors , Animals , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Rats , Rats, Sprague-Dawley , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuins/genetics , Sirtuins/metabolism
8.
ACS Med Chem Lett ; 4(10): 979-84, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24900595

ABSTRACT

Here, we describe the selection and optimization of a chemical series active in both a full-length and a fragment-based Huntington's disease (HD) assay. Twenty-four thousand small molecules were screened in a phenotypic HD assay, identifying a series of compounds bearing a 3-hydroxy-3-trifluoromethylpyrazole moiety as able to revert the toxicity induced by full-length mutant Htt by up to 50%. A chemical exploration around the series led to the identification of compound 4f, which demonstrated to be active in a Htt171-82Q rat primary striatal neuron assay and a PC12-Exon-1 based assay. This compound was selected for testing in R6/2 mice, in which it was well-tolerated and showed a positive effect on body weight and a positive trend in preventing ventricular volume enlargment. These studies provide strong rationale for further testing the potential benefits of 3-hydroxy-3-trifluoromethylpyrazoles in treating HD.

9.
J Med Chem ; 55(22): 10277-81, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23083093

ABSTRACT

α7 Nicotinic acetylcholine receptors (α7 nAChR) represent promising therapeutic candidates for the treatment of cognitive impairment associated with Alzheimer's disease (AD) and schizophrenia. A medicinal chemistry effort around previously reported compound 1 (SEN15924, WAY-361789) led to the identification of 12 (SEN78702, WYE-308775) a potent and selective full agonist of the α7 nAChR that demonstrated improved plasma stability, brain levels, and efficacy in behavioral cognition models.


Subject(s)
Brain/drug effects , Cognition/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Nicotinic Agonists/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Nicotinic/chemistry , Animals , CHO Cells , Calcium/metabolism , Chemistry, Pharmaceutical , Cricetinae , ERG1 Potassium Channel , Humans , Models, Molecular , Nicotinic Agonists/chemical synthesis , Piperidines/chemical synthesis , Pyrazoles/chemical synthesis , Rats , Receptors, Nicotinic/metabolism , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor
10.
J Med Chem ; 55(10): 4806-23, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22468936

ABSTRACT

Alpha-7 nicotinic acetylcholine receptors (α7 nAChR) are implicated in the modulation of many cognitive functions such as attention, working memory, and episodic memory. For this reason, α7 nAChR agonists represent promising therapeutic candidates for the treatment of cognitive impairment associated with Alzheimer's disease (AD) and schizophrenia. A medicinal chemistry effort, around our previously reported chemical series, permitted the discovery of a novel class of α7 nAChR agonists with improved selectivity, in particular against the α3 receptor subtype and better ADME profile. The exploration of this series led to the identification of 5-(4-acetyl[1,4]diazepan-1-yl)pentanoic acid [5-(4-methoxyphenyl)-1H-pyrazol-3-yl] amide (25, SEN15924, WAY-361789), a novel, full agonist of the α7 nAChR that was evaluated in vitro and in vivo. Compound 25 proved to be potent and selective, and it demonstrated a fair pharmacokinetic profile accompanied by efficacy in rodent behavioral cognition models (novel object recognition and auditory sensory gating).


Subject(s)
Azepines/chemical synthesis , Nicotinic Agonists/chemical synthesis , Pyrazoles/chemical synthesis , Receptors, Nicotinic/metabolism , Administration, Oral , Animals , Azepines/pharmacokinetics , Azepines/pharmacology , Brain/metabolism , Calcium/metabolism , Catalytic Domain , Cell Line , Cell Membrane Permeability , Cognition/drug effects , Dogs , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Humans , Male , Membrane Potentials/drug effects , Models, Molecular , Nicotinic Agonists/pharmacokinetics , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/chemical synthesis , Nicotinic Antagonists/pharmacokinetics , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Radioligand Assay , Rats , Rats, Long-Evans , Reflex, Startle/drug effects , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor
11.
Psychopharmacology (Berl) ; 218(4): 635-47, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21643676

ABSTRACT

RATIONALE: α7 nicotinic acetylcholine receptor (nAChR) agonists are proposed as candidate agents for the adjunctive treatment of cognitive deficits associated with schizophrenia. Despite the pursuit of such an approach clinically, it is surprising that the preclinical profile of pro-cognitive agents in conjunction with antipsychotic drugs is currently unexplored. OBJECTIVES: We determined if the memory-enhancing effects of the selective α7 nAChR agonist WYE-103914 were preserved in the presence of the atypical antipsychotic drug risperidone, and if the antipsychotic-like profile of risperidone was preserved in the presence of WYE-103914. METHODS: Using the rat novel object recognition (NOR) paradigm, the maintenance of memory-enhancing activity of the α7 nAChR agonist WYE-103914 in the presence of risperidone was examined. Similarly, in the standard tests of antipsychotic-like activity, apomorphine-induced climbing (AIC) in mice and conditioned avoidance responding (CAR) in rats, the preservation of antipsychotic-like activity of risperidone was evaluated in the presence of WYE-103914. RESULTS: WYE-103914 exhibited memory-enhancing activity in rat NOR, and this effect of WYE-103914 was retained in the presence of risperidone. In AIC, the atypical antipsychotic profile of risperidone was not significantly altered by WYE-103914. In contrast, WYE-103914 moderately potentiated the efficacy profile of risperidone in CAR, an effect that did not appear to be convincingly linked to a pharmacokinetic interaction. CONCLUSIONS: These data underscore the value of a preclinical evaluation of the adjunctive profile of a memory-enhancing agent in combination with antipsychotics and provide further support to augmentation with α7 nAChR agonists to address the cognitive deficits associated with schizophrenia.


Subject(s)
Cognition Disorders/drug therapy , Pyridines/pharmacology , Risperidone/pharmacology , Schizophrenia/drug therapy , Urea/analogs & derivatives , Animals , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Cognition Disorders/etiology , Drug Evaluation, Preclinical , Drug Interactions , Drug Therapy, Combination , Male , Memory/drug effects , Mice , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Schizophrenia/physiopathology , Urea/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
12.
FASEB J ; 25(6): 1983-2000, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21393573

ABSTRACT

Humanin (HN) is a 24-residue peptide displaying a protective activity in vitro against a range of cytotoxic and neurotoxic insults, as well as mediating in vivo amelioration of Alzheimer disease (AD)-related memory impairment in experimental models. Published evidence suggests that the mechanisms through which HN exerts its cyto- and neuroprotective activity may include its secretion and binding to membrane-associated receptors. Here, we describe the identification of a new modulator of HN neuroprotective activity, V-set and transmembrane domain containing 2 like (VSTM2L), previously known as C20orf102. VSTM2L interacts with HN in both yeast and mammalian cells, is secreted in cultured cells, is present in serum, and is selectively expressed in the central nervous system. VSTM2L colocalizes with HN in distinct brain areas as well as in primary cultured neurons, where it plays a role in the modulation of neuronal viability. When tested in HN neuroprotection bioassays, VSTM2L acts as a strong antagonist of HN neuroprotective activity. In summary, VSTM2L is the first example of a secreted antagonist of HN and may play a role in the modulation of HN biological functions.


Subject(s)
Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain/cytology , Brain/metabolism , Cell Line , Cloning, Molecular , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Saccharomyces cerevisiae , Spinal Cord/metabolism , Two-Hybrid System Techniques
13.
J Med Chem ; 53(11): 4379-89, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20465311

ABSTRACT

Alpha-7 nicotinic acetylcholine receptor (alpha7 nAChR) agonists are promising therapeutic candidates for the treatment of cognitive impairment. We report a series of novel, potent small molecule agonists (4-18) of the alpha7 nAChR deriving from our continuing efforts in the areas of Alzheimer's disease and schizophrenia. One of the compounds of the series containing a urea moiety (16) was further shown to be a selective agonist of the alpha7 nAChR with excellent in vitro and in vivo profiles, brain penetration, and oral bioavailability and demonstrated in vivo efficacy in multiple behavioral cognition models. Structural modifications leading to the improved selectivity profile and the biological evaluation of this series of compounds are discussed.


Subject(s)
Nicotinic Agonists/chemical synthesis , Nicotinic Agonists/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/pharmacology , Administration, Oral , Animals , Humans , Inhibitory Concentration 50 , Male , Models, Molecular , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacokinetics , Protein Conformation , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Rats , Receptors, Nicotinic/chemistry , Structure-Activity Relationship , Substrate Specificity , Urea/administration & dosage , Urea/pharmacokinetics , alpha7 Nicotinic Acetylcholine Receptor
14.
J Neurochem ; 112(6): 1539-51, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20050968

ABSTRACT

To investigate the role of the Wnt inhibitor Dickkopf-1 (DKK-1) in the pathophysiology of neurodegenerative diseases, we analysed DKK-1 expression and localization in transgenic mouse models expressing familial Alzheimer's disease mutations and a frontotemporal dementia mutation. A significant increase of DKK-1 expression was found in the diseased brain areas of all transgenic lines, where it co-localized with hyperphosphorylated tau-bearing neurons. In TgCRND8 mice, DKK-1 immunoreactivity was detected in neurons surrounding amyloid deposits and within the choline acetyltransferase-positive neurons of the basal forebrain. Active glycogen synthase kinase-3 (GSK-3) was found to co-localize with DKK-1 and phospho-tau staining. Downstream to GSK-3, a significant reduction in beta-catenin translocation to the nucleus, indicative of impaired Wnt signaling functions, was found as well. Cumulatively, our findings indicate that DKK-1 expression is associated with events that lead to neuronal death in neurodegenerative diseases and support a role for DKK-1 as a key mediator of neurodegeneration with therapeutic potential.


Subject(s)
Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Neurodegenerative Diseases/metabolism , Age Factors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Brain/cytology , Brain/metabolism , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/metabolism , Presenilin-1/genetics , beta Catenin/metabolism , tau Proteins/metabolism
15.
Bioorg Med Chem ; 17(14): 5247-58, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19515567

ABSTRACT

Alpha 7 nicotinic acetylcholine receptor (alpha(7) nAChR) agonists are promising therapeutic candidates for the treatment of cognitive impairment associated with a variety of disorders including Alzheimer's disease and schizophrenia. Alpha 7 nAChRs are expressed in brain regions associated with cognitive function, regulate cholinergic neurotransmission and have been shown to be down regulated in both schizophrenia and Alzheimer's disease. Herein we report a novel, potent small molecule agonist of the alpha 7 nAChR, SEN12333/WAY-317538. This compound is a selective agonist of the alpha(7) nAChR with excellent in vitro and in vivo profiles, excellent brain penetration and oral bioavailability, and demonstrates in vivo efficacy in multiple behavioural cognition models. The SAR and biological evaluation of this series of compounds are discussed.


Subject(s)
Morpholines/chemistry , Morpholines/pharmacology , Nicotinic Agonists/chemistry , Nicotinic Agonists/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , Alzheimer Disease/drug therapy , Animals , Binding, Competitive , Calcium/metabolism , Cell Line , Cognition/drug effects , Electrophysiology , Humans , Morpholines/pharmacokinetics , Nicotinic Agonists/pharmacokinetics , Pyridines/pharmacokinetics , Rats , Rats, Wistar , Schizophrenia/drug therapy , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor
16.
J Pharmacol Exp Ther ; 329(2): 459-68, 2009 May.
Article in English | MEDLINE | ID: mdl-19223665

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (nAChR) is a promising target for treatment of cognitive dysfunction associated with Alzheimer's disease and schizophrenia. Here, we report the pharmacological properties of 5-morpholin-4-yl-pentanoic acid (4-pyridin-3-yl-phenyl)-amide [SEN12333 (WAY-317538)], a novel selective agonist of alpha7 nAChR. SEN12333 shows high affinity for the rat alpha7 receptor expressed in GH4C1 cells (K(i) = 260 nM) and acts as full agonist in functional Ca(2+) flux studies (EC(50) = 1.6 microM). In whole-cell patch-clamp recordings, SEN12333 activated peak currents and maximal total charges similar to acetylcholine (EC(50) = 12 microM). The compound did not show agonist activity at other nicotinic receptors tested and acted as a weak antagonist at alpha3-containing receptors. SEN12333 treatment (3 mg/kg i.p.) improved episodic memory in a novel object recognition task in rats in conditions of spontaneous forgetting as well as cognitive disruptions induced via glutamatergic [5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate); MK-801] or cholinergic (scopolamine) mechanisms. This improvement was blocked by the alpha7-selective antagonist methyllycaconitine, indicating that it is mediated by alpha7 activation. SEN12333 also prevented a scopolamine-induced deficit in a passive avoidance task. In models targeting other cognitive domains, including attention and perceptual processing, SEN12333 normalized the apomorphine-induced deficit of prepulse inhibition. Neuroprotection of SEN12333 was demonstrated in quisqualate-lesioned animals in which treatment with SEN12333 (3 mg/kg/day i.p.) resulted in a significant protection of choline acetyltransferase-positive neurons in the lesioned hemisphere. Cumulatively, our results demonstrate that the novel alpha7 nAChR agonist SEN12333 has procognitive and neuroprotective properties, further demonstrating utility of alpha7 agonists for treatment of neurodegenerative and cognitive disorders.


Subject(s)
Cognition Disorders/drug therapy , Morpholines/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Receptors, Nicotinic/metabolism , Animals , Behavior, Animal/drug effects , Calcium/metabolism , Cell Line , Cognition/drug effects , Cognition Disorders/metabolism , Humans , Male , Membrane Potentials/drug effects , Molecular Structure , Morpholines/chemistry , Morpholines/pharmacokinetics , Morpholines/pharmacology , Motor Activity/drug effects , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Nicotinic Agonists/chemistry , Nicotinic Agonists/pharmacokinetics , Nicotinic Agonists/pharmacology , Patch-Clamp Techniques , Protein Binding , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Long-Evans , Rats, Wistar , alpha7 Nicotinic Acetylcholine Receptor
17.
Neurobiol Dis ; 24(2): 254-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16919965

ABSTRACT

Expression of the Wnt antagonist Dickkopf-1 (DKK1) is induced during neurodegenerative processes associated with Alzheimer's Disease and brain ischemia. However, little is known about DKK1-mediated effects on neurons. We now describe that, in cultured neurons, DKK1 is able to inhibit canonical Wnt signaling, as assessed by TCF reporter assay and analysis of beta-catenin levels, and to elicit cell death associated with loss of BCL-2 expression, induction of BAX, and TAU hyperphosphorylation. Local infusion of DKK1 in rats caused neuronal cell death and astrocytosis in the CA1 region of the hippocampus and death of cholinergic neurons in the nucleus basalis magnocellularis. Both effects were reversed by systemic administration of lithium ions, which rescue the Wnt pathway by inhibiting glycogen synthase kinase-3beta. The demonstration that DKK1 inhibits Wnt signaling in neurons and causes neuronal death supports the hypothesis that inhibition of the canonical Wnt pathway contributes to the pathophysiology of neurodegenerative disorders.


Subject(s)
Brain/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Wnt Proteins/metabolism , tau Proteins/metabolism , Animals , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/physiopathology , Brain/pathology , Brain/physiopathology , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/physiopathology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lithium/pharmacology , Male , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurons/pathology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction/physiology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , beta Catenin/metabolism
18.
Neurobiol Dis ; 23(2): 260-72, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16766197

ABSTRACT

In 7-month-old TgCRND8 mice, the extracellular cortical acetylcholine levels in vivo, the number and morphology of cholinergic neurons in the nucleus basalis magnocellularis and the ability to acquire an inhibitory avoidance response in the step-down test were studied. The TgCRND8 mouse brain is characterized by many beta-amyloid plaques, reduced neuronal and axonal staining, white matter demyelination, glia reaction and inducible nitric oxide synthase immunoreactivity. Choline acetyltransferase immunoreactivity in the nucleus basalis magnocellularis was significantly decreased. Basal and potassium-stimulated extracellular acetylcholine levels, investigated by microdialysis, and m2 muscarinic receptor immunoreactivity were reduced in the cortex of TgCRND8 mice, and scopolamine administration increased cortical extracellular acetylcholine levels in control but not in TgCRND8 mice. A cognitive impairment was demonstrated in the step-down test. These findings demonstrate that neuronal damage and cholinergic dysfunction in vivo underlie the impairment in learning and memory functions in this mouse model of Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Axons/physiology , Choline O-Acetyltransferase/metabolism , Neurons/physiology , Acetylcholine/metabolism , Alzheimer Disease/pathology , Animals , Coloring Agents , Humans , Indoles , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Receptor, Muscarinic M2/metabolism
19.
Proc Natl Acad Sci U S A ; 102(47): 17213-8, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16275899

ABSTRACT

Like acetylcholinesterase, butyrylcholinesterase (BChE) inactivates the neurotransmitter acetylcholine (ACh) and is hence a viable therapeutic target in Alzheimer's disease, which is characterized by a cholinergic deficit. Potent, reversible, and brain-targeted BChE inhibitors (cymserine analogs) were developed based on binding domain structures to help elucidate the role of this enzyme in the central nervous system. In rats, cymserine analogs caused long-term inhibition of brain BChE and elevated extracellular ACh levels, without inhibitory effects on acetylcholinesterase. In rat brain slices, selective BChE inhibition augmented long-term potentiation. These compounds also improved the cognitive performance (maze navigation) of aged rats. In cultured human SK-N-SH neuroblastoma cells, intra- and extracellular beta-amyloid precursor protein, and secreted beta-amyloid peptide levels were reduced without affecting cell viability. Treatment of transgenic mice that overexpressed human mutant amyloid precursor protein also resulted in lower beta-amyloid peptide brain levels than controls. Selective, reversible inhibition of brain BChE may represent a treatment for Alzheimer's disease, improving cognition and modulating neuropathological markers of the disease.


Subject(s)
Acetylcholine/metabolism , Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/drug effects , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Learning/drug effects , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/enzymology , Brain/metabolism , Cholinesterase Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/enzymology , Humans , Male , Mice , Mice, Transgenic , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Serine/administration & dosage , Serine/analogs & derivatives , Serine/pharmacology , Tumor Cells, Cultured
20.
Neurobiol Dis ; 11(2): 257-74, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12505419

ABSTRACT

Injection into the nucleus basalis of the rat of preaggregated Abeta(1-42) produced a congophylic deposit and microglial and astrocyte activation and infiltration and caused a strong inflammatory reaction characterized by IL-1beta production, increased inducible cyclooxygenase (COX-2), and inducible nitric oxide synthase (iNOS) expression. Many phospho-p38MAPK-positive cells were observed around the deposit at 7 days after Abeta injection. Phospho-p38MAPK colocalized with activated microglial cells, but not astrocytes. The inflammatory reaction was accompanied by cholinergic hypofunction. We investigated the protective effect of the selective COX-2 inhibitor rofecoxib in attenuating the inflammatory response and neurodegeneration evoked by Abeta(1-42). Rofecoxib (3 mg/kg/day, 7 days) reduced microglia and astrocyte activation, iNOS induction, and p38MAPK activation to control levels. Cholinergic hypofunction was also significantly attenuated by treatment with rofecoxib. We show here for the first time in vivo the pivotal role played by the p38MAPK microglial signal transduction pathway in the inflammatory response to the Abeta(1-42) deposit.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Basal Nucleus of Meynert/enzymology , Cholinergic Fibers/enzymology , Encephalitis/enzymology , Mitogen-Activated Protein Kinases/metabolism , Neurons/enzymology , Peptide Fragments/metabolism , Acetylcholine/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/pathology , Choline O-Acetyltransferase/drug effects , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/drug effects , Cholinergic Fibers/pathology , Cyclooxygenase 2 , Down-Regulation/drug effects , Down-Regulation/physiology , Encephalitis/chemically induced , Encephalitis/physiopathology , Gliosis/chemically induced , Immunohistochemistry , Interleukin-1/metabolism , Isoenzymes/drug effects , Isoenzymes/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinases/drug effects , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Peptide Fragments/pharmacology , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...