Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(13): 11168-11181, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38932616

ABSTRACT

ß-Glucocerebrosidase (GBA/GCase) mutations leading to misfolded protein cause Gaucher's disease and are a major genetic risk factor for Parkinson's disease and dementia with Lewy bodies. The identification of small molecule pharmacological chaperones that can stabilize the misfolded protein and increase delivery of degradation-prone mutant GCase to the lysosome is a strategy under active investigation. Here, we describe the first use of fragment-based drug discovery (FBDD) to identify pharmacological chaperones of GCase. The fragment hits were identified by using X-ray crystallography and biophysical techniques. This work led to the discovery of a series of compounds that bind GCase with nM potency and positively modulate GCase activity in cells.


Subject(s)
Allosteric Site , Drug Discovery , Glucosylceramidase , Glucosylceramidase/metabolism , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/chemistry , Humans , Crystallography, X-Ray , Structure-Activity Relationship , Models, Molecular , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/metabolism
2.
ACS Med Chem Lett ; 15(5): 646-652, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38746889

ABSTRACT

The potassium (K+) ion channel KCNK13 is specifically expressed in human microglia with elevated expression observed in post-mortem human brain tissue from patients with Alzheimer's disease. Modulation of KCNK13 activity by a small-molecule inhibitor is proposed as a potential treatment for neurodegenerative diseases. Herein, we describe the evolution of a series of KCNK13 inhibitors derived from a high-throughput screening campaign, resulting in CVN293, a potent, selective, and brain permeable clinical candidate molecule. CVN293 demonstrated a concentration-dependent inhibition of the NLRP3-inflammasome mediated production of IL-1ß from LPS-primed murine microglia. Cross-species pharmacokinetic data of CVN293 are also disclosed. These findings support the advancement of CVN293 in clinical trials.

3.
J Neuroinflammation ; 21(1): 58, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409076

ABSTRACT

Neuroinflammation is highly influenced by microglia, particularly through activation of the NLRP3 inflammasome and subsequent release of IL-1ß. Extracellular ATP is a strong activator of NLRP3 by inducing K+ efflux as a key signaling event, suggesting that K+-permeable ion channels could have high therapeutic potential. In microglia, these include ATP-gated THIK-1 K+ channels and P2X7 receptors, but their interactions and potential therapeutic role in the human brain are unknown. Using a novel specific inhibitor of THIK-1 in combination with patch-clamp electrophysiology in slices of human neocortex, we found that THIK-1 generated the main tonic K+ conductance in microglia that sets the resting membrane potential. Extracellular ATP stimulated K+ efflux in a concentration-dependent manner only via P2X7 and metabotropic potentiation of THIK-1. We further demonstrated that activation of P2X7 was mandatory for ATP-evoked IL-1ß release, which was strongly suppressed by blocking THIK-1. Surprisingly, THIK-1 contributed only marginally to the total K+ conductance in the presence of ATP, which was dominated by P2X7. This suggests a previously unknown, K+-independent mechanism of THIK-1 for NLRP3 activation. Nuclear sequencing revealed almost selective expression of THIK-1 in human brain microglia, while P2X7 had a much broader expression. Thus, inhibition of THIK-1 could be an effective and, in contrast to P2X7, microglia-specific therapeutic strategy to contain neuroinflammation.


Subject(s)
Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases , Ion Channels/metabolism , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/metabolism , Receptors, Purinergic P2X7/metabolism
4.
Bioorg Med Chem Lett ; 100: 129629, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38295907

ABSTRACT

Modulators of orexin receptors are being developed for neurological illnesses such as sleep disorders, addictive behaviours and other psychiatric diseases. We herein describe the discovery of CVN766, a potent orexin 1 receptor antagonist that has greater than 1000-fold selectivity for the orexin 1 receptor over the orexin 2 receptor and demonstrates low off target hits in a diversity screen. In agreement with its in vitro ADME data, CVN766 demonstrated moderate in vivo clearance in rodents and displayed good brain permeability and target occupancy. This drug candidate is currently being investigated in clinical trials for schizophrenia and related psychiatric conditions.


Subject(s)
Disclosure , Mental Disorders , Humans , Orexins , Orexin Receptor Antagonists/pharmacology , Orexin Receptors
5.
J Med Chem ; 66(18): 12858-12876, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37708305

ABSTRACT

From our NETSseq-derived human brain transcriptomics data, we identified GPR55 as a potential molecular target for the treatment of motor symptoms in patients with Parkinson's disease. From a high-throughput screen, we identified and optimized agonists with nanomolar potency against both human and rat GPR55. We discovered compounds with either strong or limited ß-arrestin signaling and receptor desensitization, indicating biased signaling. A compound that showed minimal GPR55 desensitization demonstrated a reduction in firing frequency of medium spiny neurons cultured from rat striatum but did not reverse motor deficits in a rat hypolocomotion model. Further profiling of several desensitizing and non-desensitizing lead compounds showed that they are selective over related cannabinoid receptors CB1 and CB2 and that unbound brain concentrations well above the respective GPR55 EC50 can be readily achieved following oral administration. The novel brain-penetrant GPR55 agonists disclosed can be used to probe the role of this receptor in the brain.


Subject(s)
Cannabinoid Receptor Agonists , Signal Transduction , Humans , Rats , Animals , Receptors, Cannabinoid , beta-Arrestins , Corpus Striatum/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptor, Cannabinoid, CB2 , Receptor, Cannabinoid, CB1
6.
J Med Chem ; 66(17): 11718-11731, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37651656

ABSTRACT

Nicotinic acetylcholine receptor (nAChR) α6 subunit RNA expression is relatively restricted to midbrain regions and is located presynaptically on dopaminergic neurons projecting to the striatum. This subunit modulates dopamine neurotransmission and may have therapeutic potential in movement disorders. We aimed to develop potent and selective α6-containing nAChR antagonists to explore modulation of dopamine release and regulation of motor function in vivo. High-throughput screening (HTS) identified novel α6-containing nAChR antagonists and led to the development of CVN417. This molecule blocks α6-containing nAChR activity in recombinant cells and reduces firing frequency of noradrenergic neurons in the rodent locus coeruleus. CVN417 modulated phasic dopaminergic neurotransmission in an impulse-dependent manner. In a rodent model of resting tremor, CVN417 attenuated this behavioral phenotype. These data suggest that selective antagonism of α6-containing nAChR, with molecules such as CVN417, may have therapeutic utility in treating the movement dysfunctions observed in conditions such as Parkinson's disease.


Subject(s)
Dopamine , Receptors, Nicotinic , Brain , Cell Membrane , Corpus Striatum , Nicotinic Antagonists/pharmacology
7.
ACS Med Chem Lett ; 14(4): 442-449, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37077399

ABSTRACT

The low affinity metabotropic glutamate receptor mGluR7 has been implicated in numerous CNS disorders; however, a paucity of potent and selective activators has hampered full delineation of the functional role and therapeutic potential of this receptor. In this work, we present the identification, optimization, and characterization of highly potent, novel mGluR7 agonists. Of particular interest is the chromane CVN636, a potent (EC50 7 nM) allosteric agonist which demonstrates exquisite selectivity for mGluR7 compared to not only other mGluRs, but also a broad range of targets. CVN636 demonstrated CNS penetrance and efficacy in an in vivo rodent model of alcohol use disorder. CVN636 thus has potential to progress as a drug candidate in CNS disorders involving mGluR7 and glutamatergic dysfunction.

8.
Neuropharmacology ; 224: 109330, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36375694

ABSTRACT

Neuroinflammation, specifically the NLRP3 inflammasome cascade, is a common underlying pathological feature of many neurodegenerative diseases. Evidence suggests that NLRP3 activation involves changes in intracellular K+. Nuclear Enriched Transcript Sort Sequencing (NETSseq), which allows for deep sequencing of purified cell types from human post-mortem brain tissue, demonstrated a highly specific expression of the tandem pore domain halothane-inhibited K+ channel 1 (THIK-1) in microglia compared to other glial and neuronal cell types in the human brain. NETSseq also showed a significant increase of THIK-1 in microglia isolated from cortical regions of brains with Alzheimer's disease (AD) relative to control donors. Herein, we report the discovery and pharmacological characterisation of C101248, the first selective small-molecule inhibitor of THIK-1. C101248 showed a concentration-dependent inhibition of both mouse and human THIK-1 (IC50: ∼50 nM) and was inactive against K2P family members TREK-1 and TWIK-2, and Kv2.1. Whole-cell patch-clamp recordings of microglia from mouse hippocampal slices showed that C101248 potently blocked both tonic and ATP-evoked THIK-1 K+ currents. Notably, C101248 had no effect on other constitutively active resting conductance in slices from THIK-1-depleted mice. In isolated microglia, C101248 prevented NLRP3-dependent release of IL-1ß, an effect not seen in THIK-1-depleted microglia. In conclusion, we demonstrated that inhibiting THIK-1 (a microglia specific gene that is upregulated in brains from donors with AD) using a novel selective modulator attenuates the NLRP3-dependent release of IL-1ß from microglia, which suggests that this channel may be a potential therapeutic target for the modulation of neuroinflammation in AD.


Subject(s)
Alzheimer Disease , Inflammasomes , Potassium Channels, Tandem Pore Domain , Animals , Humans , Mice , Alzheimer Disease/metabolism , Brain/metabolism , Inflammasomes/metabolism , Microglia , Neuroinflammatory Diseases , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
9.
Glia ; 70(7): 1301-1316, 2022 07.
Article in English | MEDLINE | ID: mdl-35353387

ABSTRACT

The NLRP3 (NLR family, pyrin domain containing 3) inflammasome is a multi-protein complex responsible for the activation of caspase-1 and the subsequent cleavage and activation of the potent proinflammatory cytokines IL-1ß and IL-18, and pyroptotic cell death. NLRP3 is implicated as a driver of inflammation in a range of disorders including neurodegenerative diseases, type 2 diabetes, and atherosclerosis. A commonly reported mechanism contributing to NLRP3 inflammasome activation is potassium ion (K+ ) efflux across the plasma membrane. Identification of K+ channels involved in NLRP3 activation remains incomplete. Here, we investigated the role of the K+ channel THIK-1 in NLRP3 activation. Both pharmacological inhibitors and cells from THIK-1 knockout (KO) mice were used to assess THIK-1 contribution to macrophage NLRP3 activation in vitro. Pharmacological inhibition of THIK-1 inhibited caspase-1 activation and IL-1ß release from mouse bone-marrow-derived macrophages (BMDMs), mixed glia, and microglia in response to NLRP3 agonists. Similarly, BMDMs and microglia from THIK-1 KO mice had reduced NLRP3-dependent IL-1ß release in response to P2X7 receptor activation with ATP. Overall, these data suggest that THIK-1 is a regulator of NLRP3 inflammasome activation in response to ATP and identify THIK-1 as a potential therapeutic target for inflammatory disease.


Subject(s)
Diabetes Mellitus, Type 2 , Inflammasomes , Potassium Channels, Tandem Pore Domain/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspase 1/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism , Potassium Channels
10.
Bioorg Med Chem Lett ; 61: 128607, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35123006

ABSTRACT

We report a significant decrease in transcription of the G protein-coupled receptor GPR39 in striatal neurons of Parkinson's disease patients compared to healthy controls, suggesting that a positive modulator of GPR39 may beneficially impact neuroprotection. To test this notion, we developed various structurally diverse tool molecules. While we elaborated on previously reported starting points, we also performed an in silico screen which led to completely novel pharmacophores. In vitro studies indicated that GPR39 agonism does not have a profound effect on neuroprotection.


Subject(s)
Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Allosteric Regulation/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
11.
J Pharmacol Exp Ther ; 377(3): 407-416, 2021 06.
Article in English | MEDLINE | ID: mdl-33795395

ABSTRACT

GPR6 is an orphan G-protein-coupled receptor that has enriched expression in the striatopallidal, indirect pathway and medium spiny neurons of the striatum. This pathway is greatly impacted by the loss of the nigro-striatal dopaminergic neurons in Parkinson disease, and modulating this neurocircuitry can be therapeutically beneficial. In this study, we describe the in vitro and in vivo pharmacological characterization of (R)-1-(2-(4-(2,4-difluorophenoxy)piperidin-1-yl)-3-((tetrahydrofuran-3-yl)amino)-7,8-dihydropyrido[3,4-b]pyrazin-6(5H)-yl)ethan-1-one (CVN424), a highly potent and selective small-molecule inverse agonist for GPR6 that is currently undergoing clinical evaluation. CVN424 is brain-penetrant and shows dose-dependent receptor occupancy that attained brain 50% of receptor occupancy at plasma concentrations of 6.0 and 7.4 ng/ml in mice and rats, respectively. Oral administration of CVN424 dose-dependently increases locomotor activity and reverses haloperidol-induced catalepsy. Furthermore, CVN424 restored mobility in bilateral 6-hydroxydopamine lesion model of Parkinson disease. The presence and localization of GPR6 in medium spiny neurons of striatum postmortem samples from both nondemented control and patients with Parkinson disease were confirmed at the level of both RNA (using Nuclear Enriched Transcript Sort sequencing) and protein. This body of work demonstrates that CVN424 is a potent, orally active, and brain-penetrant GPR6 inverse agonist that is effective in preclinical models and is a potential therapeutic for improving motor function in patients with Parkinson disease. SIGNIFICANCE STATEMENT: CVN424 represents a nondopaminergic novel drug for potential use in patients with Parkinson disease.


Subject(s)
Corpus Striatum , Animals , Gonadal Steroid Hormones , Rats
12.
Stem Cell Reports ; 16(5): 1276-1289, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891871

ABSTRACT

Sequestosome-1 (SQSTM1/p62) is involved in cellular processes such as autophagy and metabolic reprogramming. Mutations resulting in the loss of function of SQSTM1 lead to neurodegenerative diseases including frontotemporal dementia. The pathogenic mechanism that contributes to SQSTM1-related neurodegeneration has been linked to its role as an autophagy adaptor, but this is poorly understood, and its precise role in mitochondrial function and clearance remains to be clarified. Here, we assessed the importance of SQSTM1 in human induced pluripotent stem cell (iPSC)-derived cortical neurons through the knockout of SQSTM1. We show that SQSTM1 depletion causes altered mitochondrial gene expression and functionality, as well as autophagy flux, in iPSC-derived neurons. However, SQSTM1 is not essential for mitophagy despite having a significant impact on early PINK1-dependent mitophagy processes including PINK1 recruitment and phosphorylation of ubiquitin on depolarized mitochondria. These findings suggest that SQSTM1 is important for mitochondrial function rather than clearance.


Subject(s)
Cerebral Cortex/cytology , Mitochondria/metabolism , Neurons/metabolism , Sequestosome-1 Protein/metabolism , Cell Differentiation , Cell Respiration , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial , Mitochondria/pathology , Mitophagy , Oxidative Phosphorylation , Protein Kinases/metabolism , Reproducibility of Results
13.
Front Aging Neurosci ; 12: 242, 2020.
Article in English | MEDLINE | ID: mdl-33117143

ABSTRACT

Kinases are an intensively studied drug target class in current pharmacological research as evidenced by the large number of kinase inhibitors being assessed in clinical trials. Kinase-targeted therapies have potential for treatment of a broad array of indications including central nervous system (CNS) disorders. In addition to the many variables which contribute to identification of a successful therapeutic molecule, drug discovery for CNS-related disorders also requires significant consideration of access to the target organ and specifically crossing the blood-brain barrier (BBB). To date, only a small number of kinase inhibitors have been reported that are specifically designed to be BBB permeable, which nonetheless demonstrates the potential for success. This review considers the potential for kinase inhibitors in the context of unmet medical need for neurodegenerative disease. A subset of kinases that have been the focus of clinical investigations over a 10-year period have been identified and discussed individually. For each kinase target, the data underpinning the validity of each in the context of neurodegenerative disease is critically evaluated. Selected molecules for each kinase are identified with information on modality, binding site and CNS penetrance, if known. Current clinical development in neurodegenerative disease are summarized. Collectively, the review indicates that kinase targets with sufficient rationale warrant careful design approaches with an emphasis on improving brain penetrance and selectivity.

14.
Front Immunol ; 11: 293, 2020.
Article in English | MEDLINE | ID: mdl-32194553

ABSTRACT

Inflammation is considered a mechanistic driver of Alzheimer's disease, thought to increase tau phosphorylation, the first step to the formation of neurofibrillary tangles (NFTs). To further understand how inflammation impacts the development of tau pathology, we used (hTau) mice, which express all six, non-mutated, human tau isoforms, but with an altered ratio of tau isoforms favoring 3R tau due to the concomitant loss of murine tau (mTau) that is predominantly 4R. Such an imbalance pattern has been related to susceptibility to NFTs formation, but whether or not this also affects susceptibility to systemic inflammation and related changes in tau phosphorylation is not known. To reduce the predominance of 3R tau by increasing 4R tau availability, we bred hTau mice on a heterozygous mTau background and compared the impact of systemic inflammation induced by lipopolysaccharide (LPS) in hTau mice hetero- or homozygous mTau knockout. Three-month-old male wild-type (Wt), mTau+/-, mTau-/-, hTau/mTau+/-, and hTau/mTau-/- mice were administered 100, 250, or 330 µg/kg of LPS or its vehicle phosphate buffer saline (PBS) [intravenously (i.v.), n = 8-9/group]. Sickness behavior, reflected by behavioral suppression in the spontaneous alternation task, hippocampal tau phosphorylation, measured by western immunoblotting, and circulating cytokine levels were quantified 4 h after LPS administration. The persistence of the LPS effects (250 µg/kg) on these measures, and food burrowing behavior, was assessed at 24 h post-inoculation in Wt, mTau+/-, and hTau/mTau+/- mice (n = 9-10/group). In the absence of immune stimulation, increasing 4R tau levels in hTau/mTau+/- exacerbated pS202 and pS396/404 tau phosphorylation, without altering total tau levels or worsening early behavioral perturbations characteristic of hTau/mTau-/- mice. We also show for the first time that modulating 4R tau levels in hTau mice affects the response to systemic inflammation. Behavior was suppressed in all genotypes 4 h following LPS administration, but hTau/mTau+/- exhibited more severe sickness behavior at the 100 µg/kg dose and a milder behavioral and cytokine response than hTau/mTau-/- mice at the 330 µg/kg dose. All LPS doses decreased tau phosphorylation at both epitopes in hTau/mTau+/- mice, but pS202 levels were selectively reduced at the 100 µg/kg dose in hTau/mTau-/- mice. Behavioral suppression and decreased tau phosphorylation persisted at 24 h following LPS administration in hTau/mTau+/- mice.


Subject(s)
Hippocampus/metabolism , Inflammation/complications , Tauopathies/etiology , tau Proteins/metabolism , Animals , Cytokines/biosynthesis , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Isoforms/analysis , tau Proteins/analysis
15.
Alzheimers Dement (N Y) ; 5: 241-253, 2019.
Article in English | MEDLINE | ID: mdl-31297438

ABSTRACT

Since the G8 dementia summit in 2013, a number of initiatives have been established with the aim of facilitating the discovery of a disease-modifying treatment for dementia by 2025. This report is a summary of the findings and recommendations of a meeting titled "Tackling gaps in developing life-changing treatments for dementia", hosted by Alzheimer's Research UK in May 2018. The aim of the meeting was to identify, review, and highlight the areas in dementia research that are not currently being addressed by existing initiatives. It reflects the views of leading experts in the field of neurodegeneration research challenged with developing a strategic action plan to address these gaps and make recommendations on how to achieve the G8 dementia summit goals. The plan calls for significant advances in (1) translating newly identified genetic risk factors into a better understanding of the impacted biological processes; (2) enhanced understanding of selective neuronal resilience to inform novel drug targets; (3) facilitating robust and reproducible drug-target validation; (4) appropriate and evidence-based selection of appropriate subjects for proof-of-concept clinical trials; (5) improving approaches to assess drug-target engagement in humans; and (6) innovative approaches in conducting clinical trials if we are able to detect disease 10-15 years earlier than we currently do today.

16.
Sci Rep ; 8(1): 5667, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618797

ABSTRACT

Inflammation is an established contributor to disease and the NLRP3 inflammasome is emerging as a potential therapeutic target. A number of small molecule inhibitors of the NLRP3 pathway have been described. Here we analysed the most promising of these inhibitor classes side by side to assess relative potency and selectivity for their respective putative targets. Assessed using ASC inflammasome-speck formation, and release of IL-1ß, in both human monocyte/macrophage THP1 cells and in primary mouse microglia, we compared the relative potency and selectivity of P2X7 inhibitors, inflammasome inhibitors (diarylsulfonylurea vs. the NBC series), and caspase-1 inhibitors. In doing so we are now able to provide a well characterised small molecule tool kit for interrogating and validating inflammasome-dependent responses with a range of nanomolar potency inhibitors against established points in the inflammasome pathway.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Macrophages/immunology , Microglia/immunology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Animals, Newborn , Cells, Cultured , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Monocytes/cytology , Monocytes/metabolism , Signal Transduction
17.
Exp Gerontol ; 94: 103-107, 2017 08.
Article in English | MEDLINE | ID: mdl-27979768

ABSTRACT

Alzheimer's disease (AD), the predominant form of dementia, is highly correlated with the abnormal hyperphosphorylation and aggregation of tau. Immune responses are key drivers of AD and how they contribute to tau pathology in human disease remains largely unknown. This review summarises current knowledge on the association between inflammatory processes and tau pathology. While, preclinical evidence suggests that inflammation can indeed induce tau hyperphosphorylation at both pre- and post-tangles epitopes, a better understanding of whether this develops into advanced pathological features such as neurofibrillary tangles is needed. Microglial cells, the immune phagocytes in the central nervous system, appear to play a key role in regulating tau pathology, but the underlying mechanisms are not fully understood. Their activation can be detrimental via the secretion of pro-inflammatory mediators, particularly interleukin-1ß, but also potentially beneficial through phagocytosis of extracellular toxic tau oligomers. Nevertheless, anti-inflammatory treatments in animal models were found protective, but whether or not they affect microglial phagocytosis of tau species is unknown. However, one major challenge to our understanding of the role of inflammation in the progression of tau pathology is the preclinical models used to address this question. They mostly rely on the use of septic doses of lipopolysaccharide that do not reflect the inflammatory conditions experienced AD patients, questioning whether the impact of inflammation on tau pathology in these models is dose-dependent and relevant to the human disease. The use of more translational models of inflammation corroborated with verification in clinical investigations are necessary to progress our understanding of the interplay between inflammation and tau pathology.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Protein Aggregation, Pathological , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Anti-Inflammatory Agents/therapeutic use , Brain/drug effects , Brain/pathology , Brain/physiopathology , Humans , Inflammation/drug therapy , Inflammation/pathology , Inflammation/physiopathology , Neuroprotective Agents/therapeutic use , Phosphorylation , Signal Transduction
18.
Br J Pharmacol ; 174(5): 370-385, 2017 03.
Article in English | MEDLINE | ID: mdl-28009436

ABSTRACT

BACKGROUND AND PURPOSE: AMPA receptor positive allosteric modulators represent a potential therapeutic strategy to improve cognition in people with schizophrenia. These studies collectively constitute the preclinical pharmacology data package used to build confidence in the pharmacology of this molecule and enable a clinical trial application. EXPERIMENTAL APPROACH: [N-[(2S)-5-(6-fluoro-3-pyridinyl)-2,3-dihydro 1H-inden-2-yl]-2-propanesulfonamide] (UoS12258) was profiled in a number of in vitro and in vivo studies to highlight its suitability as a novel therapeutic agent. KEY RESULTS: We demonstrated that UoS12258 is a selective, positive allosteric modulator of the AMPA receptor. At rat native hetero-oligomeric AMPA receptors, UoS12258 displayed a minimum effective concentration of approximately 10 nM in vitro and enhanced AMPA receptor-mediated synaptic transmission at an estimated free brain concentration of approximately 15 nM in vivo. UoS12258 reversed a delay-induced deficit in novel object recognition in rats after both acute and sub-chronic dosing. Sub-chronic dosing reduced the minimum effective dose from 0.3 to 0.03 mg·kg-1 . UoS12258 was also effective at improving performance in two other cognition models, passive avoidance in scopolamine-impaired rats and water maze learning and retention in aged rats. In side-effect profiling studies, UoS12258 did not produce significant changes in the maximal electroshock threshold test at doses below 10 mg·kg-1 . CONCLUSION AND IMPLICATIONS: We conclude that UoS12258 is a potent and selective AMPA receptor modulator exhibiting cognition enhancing properties in several rat behavioural models superior to other molecules that have previously entered clinical evaluation.


Subject(s)
Behavior, Animal/drug effects , Indenes/pharmacology , Nootropic Agents/pharmacology , Receptors, AMPA/drug effects , Sulfonamides/pharmacology , Allosteric Regulation/drug effects , Animals , Avoidance Learning/drug effects , Cognition/drug effects , Dose-Response Relationship, Drug , Electroshock , Humans , Indenes/administration & dosage , Indenes/toxicity , Male , Maze Learning/drug effects , Nootropic Agents/administration & dosage , Nootropic Agents/toxicity , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, AMPA/metabolism , Recognition, Psychology/drug effects , Scopolamine/toxicity , Sulfonamides/administration & dosage , Sulfonamides/toxicity
19.
Bioorg Med Chem Lett ; 25(24): 5792-6, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26531152

ABSTRACT

Herein we describe a series of tetrahydrobenzotriazoles as novel, potent metabotropic glutamate receptor subtype 5 (mGlu5) positive allosteric modulators (PAMs). Exploration of the SAR surrounding the tetrahydrobenzotriazole core ultimately led to the identification of 29 as a potent mGlu5 PAM with a low maximal glutamate potency fold shift, acceptable in vitro DMPK parameters and in vivo PK profile and efficacy in the rat novel object recognition (NOR) assay. As a result 29 was identified as a suitable compound for progression to in vivo safety evaluation.


Subject(s)
Antipsychotic Agents/chemistry , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Triazoles/chemistry , Allosteric Regulation/drug effects , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cognition/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Humans , Microsomes/metabolism , Rats , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship , Triazoles/metabolism , Triazoles/pharmacology
20.
Expert Opin Drug Discov ; 8(12): 1529-39, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24195711

ABSTRACT

INTRODUCTION: Vilazodone is the newest serotonergic antidepressant to be approved by the FDA for the treatment of major depressive disorder (MDD). Vilazodone is a combined serotonin specific reuptake inhibitor (SSRI) and 5-HT(1A) receptor partial agonist. It was originally designed based on the premise that negative feedback circuitry mediated through somatodendritic 5-HT1 autoreceptors, limits the acute SSRI-induced enhancements in serotonergic neurotransmission. Therefore, the combination of SSRI with 5-HT(1A) receptor agonism should temporally enhance the neuroplastic adaptation and subsequently hasten therapeutic efficacy compared to current treatments. AREAS COVERED: This review provides the history and preclinical development of vilazodone, highlighting the available data on its putative mechanism of action, potential clinical profile and possible areas for differentiation. These preclinical hypotheses will be contextualised with an overview of the key findings from the current clinic data on vilazodone. EXPERT OPINION: Preclinical data packages on vilazodone have clearly demonstrated its SSRI activity. In isolated in vitro systems, 5-HT(1A) receptor agonism has been demonstrated, but the recapitulation of this activity in vivo has been inconclusive. This uncertainty of its in vivo profile has largely translated to the clinical scenario with efficacy and adverse event profiles being similar to that seen with SSRIs in MDD. More in-depth evaluation of the two Phase III studies have also provided some early evidence of differentiation on onset of therapeutic benefit, anxiety measures and improvements in sexual function. Further evaluation of MDD and anxiety patient outcomes is essential to demonstrate if vilazodone is truly a novel therapeutic.


Subject(s)
Antidepressive Agents/therapeutic use , Benzofurans/therapeutic use , Depression/drug therapy , Indoles/therapeutic use , Piperazines/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Animals , Antidepressive Agents/pharmacology , Benzofurans/pharmacology , Drug Discovery , Humans , Indoles/pharmacology , Piperazines/pharmacology , Vilazodone Hydrochloride
SELECTION OF CITATIONS
SEARCH DETAIL
...