Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Neurosci ; 42(44): 8225-8236, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36163142

ABSTRACT

Phosphodiesterase (PDE) inhibitors have been safely and effectively used in the clinic and increase the concentration of intracellular cyclic nucleotides (cAMP/cGMP). These molecules activate downstream mediators, including the cAMP response element-binding protein (CREB), which controls neuronal excitability and growth responses. CREB gain of function enhances learning and allocates neurons into memory engrams. CREB also controls recovery after stroke. PDE inhibitors are linked to recovery from neural damage and to stroke recovery in specific sites within the brain. PDE2A is enriched in cortex. In the present study, we use a mouse cortical stroke model in young adult and aged male mice to test the effect of PDE2A inhibition on functional recovery, and on downstream mechanisms of axonal sprouting, tissue repair, and the functional connectivity of neurons in recovering cortex. Stroke causes deficits in use of the contralateral forelimb, loss of axonal projections in cortex adjacent to the infarct, and functional disconnection of neuronal networks. PDE2A inhibition enhances functional recovery, increases axonal projections in peri-infarct cortex, and, through two-photon in vivo imaging, enhances the functional connectivity of motor system excitatory neurons. PDE2A inhibition after stroke does not have an effect on other aspects of tissue repair, such as angiogenesis, gliogenesis, neurogenesis, and inflammatory responses. These data suggest that PDE2A inhibition is an effective therapeutic approach for stroke recovery in the rodent and that it simultaneously enhances connectivity in peri-infarct neuronal populations.SIGNIFICANCE STATEMENT Inhibition of PDE2A enhances motor recovery, axonal projections, and functional connectivity of neurons in peri-infarct tissue. This represents an avenue for a pharmacological therapy for stroke recovery.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2 , Stroke , Animals , Male , Mice , Cyclic AMP Response Element-Binding Protein , Infarction , Motor Neurons , Neurogenesis , Phosphodiesterase Inhibitors/pharmacology , Recovery of Function/physiology , Stroke/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors
2.
J Pharmacol Exp Ther ; 365(1): 179-188, 2018 04.
Article in English | MEDLINE | ID: mdl-29440309

ABSTRACT

The pathophysiology of schizophrenia has been associated with glutamatergic dysfunction. Modulation of the glutamatergic signaling pathway, including N-methyl-d-aspartate (NMDA) receptors, can provide a new therapeutic target for schizophrenia. Phosphodiesterase 2A (PDE2A) is highly expressed in the forebrain, and is a dual substrate enzyme that hydrolyzes both cAMP and cGMP, which play pivotal roles as intracellular second messengers downstream of NMDA receptors. Here we characterize the in vivo pharmacological profile of a selective and brain-penetrant PDE2A inhibitor, (N-{(1S)-1-[3-fluoro-4-(trifluoromethoxy)phenyl]-2-methoxyethyl}-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide) (TAK-915) as a novel treatment of schizophrenia. Oral administration of TAK-915 at 3 and 10 mg/kg significantly increased cGMP levels in the frontal cortex, hippocampus, and striatum of rats. TAK-915 at 10 mg/kg significantly upregulated the phosphorylation of α-amino-3-hydroxy-5-methylisoxazole-4-proprionic acid receptor subunit GluR1 in the rat hippocampus. TAK-915 at 3 and 10 mg/kg significantly attenuated episodic memory deficits induced by the NMDA receptor antagonist (+)-MK-801 hydrogen maleate (MK-801) in the rat passive avoidance test. TAK-915 at 10 mg/kg significantly attenuated working memory deficits induced by MK-801 in the rat radial arm maze test. Additionally, TAK-915 at 10 mg/kg prevented subchronic phencyclidine-induced social withdrawal in social interaction in rats. In contrast, TAK-915 did not produce antipsychotic-like activity; TAK-915 had little effect on MK-801- or methamphetamine-induced hyperlocomotion in rats. These results suggest that TAK-915 has a potential to ameliorate cognitive impairments and social withdrawal in schizophrenia.


Subject(s)
Cognitive Dysfunction/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Pyrazines/pharmacology , Pyridines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/complications , Social Behavior , Animals , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Avoidance Learning/drug effects , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Locomotion/drug effects , Male , Memory, Episodic , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphodiesterase Inhibitors/therapeutic use , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Receptors, AMPA/metabolism , Schizophrenia/chemically induced
3.
Chem Pharm Bull (Tokyo) ; 65(11): 1058-1077, 2017.
Article in English | MEDLINE | ID: mdl-29093293

ABSTRACT

It has been hypothesized that selective inhibition of phosphodiesterase (PDE) 2A could potentially be a novel approach to treat cognitive impairment in neuropsychiatric and neurodegenerative disorders through augmentation of cyclic nucleotide signaling pathways in brain regions associated with learning and memory. Following our earlier work, this article describes a drug design strategy for a new series of lead compounds structurally distinct from our clinical candidate 2 (TAK-915), and subsequent medicinal chemistry efforts to optimize potency, selectivity over other PDE families, and other preclinical properties including in vitro phototoxicity and in vivo rat plasma clearance. These efforts resulted in the discovery of N-((1S)-2-hydroxy-2-methyl-1-(4-(trifluoromethoxy)phenyl)propyl)-6-methyl-5-(3-methyl-1H-1,2,4-triazol-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (20), which robustly increased 3',5'-cyclic guanosine monophosphate (cGMP) levels in the rat brain following an oral dose, and moreover, attenuated MK-801-induced episodic memory deficits in a passive avoidance task in rats. These data provide further support to the potential therapeutic utility of PDE2A inhibitors in enhancing cognitive performance.


Subject(s)
Cognition Disorders/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Discovery , Phosphodiesterase Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , 3T3 Cells , Administration, Oral , Animals , COS Cells , Cell Survival/drug effects , Chlorocebus aethiops , Cognition Disorders/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Molecular Structure , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry , Powder Diffraction , Pyrazines/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Rats , Rats, Long-Evans , Solubility , Structure-Activity Relationship , Thermodynamics
4.
Respir Res ; 13: 55, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22731784

ABSTRACT

BACKGROUND: The mechanisms underlying the association between smoking and mucus overproduction remain unknown. Because of its involvement in other airway diseases, such as asthma, we hypothesized that CaĀ²Ć¢ĀĀŗ-activated ClĆ¢ĀĀ» channel 1 (CLCA1) was associated with overproduction of mucus in the airways of smokers and COPD patients. METHODS: Using real-time quantitative PCR analyses, we compared the CLCA1 mRNA expression levels in induced-sputum cells from COPD patients (n = 20), smokers without COPD (n = 5), and non-smokers (n =13). We also examined the relationship between CLCA1 protein expression and mucus production in lung airway epithelia of COPD patients (n = 6), smokers without COPD (n = 7), and non-smokers (n = 7). RESULTS: CLCA1 mRNA expression was significantly up-regulated in the induced-sputum cells of COPD patients compared with cells of non-smokers (p = 0.02), but there was no significant difference compared with cells of smokers without COPD. Using immunostaining with an anti-CLCA1 antibody, semi-quantitative image analyses of airway epithelium demonstrated significantly increased CLCA1 expression in smokers without COPD (p = 0.02) and in COPD patients (p = 0.002) compared with non-smokers. There were significant negative correlations between CLCA1 protein expression and FEV1/FVC (r = -0.57, p = 0.01) and %predicted FEV1 (r = -0.56, p = 0.01). PAS staining for mucus showed that there was a significant positive correlation between CLCA1 protein expression and mucus production (r = 0.67, p = 0.001). These markers were significantly increased in smokers without COPD (p = 0.04) and in COPD patients (p = 0.003) compared with non-smokers (non-smokers < smokers ≤ COPD). CONCLUSIONS: CLCA1 expression is significantly related to mucus production in the airway epithelia of smokers and COPD patients, and may contribute to the development and pathogenesis of COPD by inducing mucus production.


Subject(s)
Chloride Channels/metabolism , Epithelial Cells/metabolism , Lung/metabolism , Mucus/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism , Smoking/adverse effects , Adult , Aged , Case-Control Studies , Chloride Channels/genetics , Female , Forced Expiratory Volume , Humans , Immunohistochemistry , Interleukin-13 Receptor alpha1 Subunit , Lung/physiopathology , Male , Middle Aged , Mucin 5AC/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-13/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sputum/cytology , Up-Regulation , Vital Capacity
5.
J Neurochem ; 119(6): 1330-40, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21992552

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder leading to a progressive loss of cognitive function and is pathologically characterized by senile plaques and neurofibrillary tangles. Glycogen synthase kinase-3 (GSK-3) is involved in AD pathogenesis. GSK-3 is reported not only to phosphorylate tau, a major component of neurofibrillary tangles, but also to regulate the production of amyloid Ɵ, which is deposited in senile plaques. Therefore, pharmacological inhibition of GSK-3 is considered an attractive therapeutic approach. In this study, we report the pharmacological effects of a novel GSK-3 inhibitor, 2-methyl-5-(3-{4-[(S)-methylsulfinyl]phenyl}-1-benzofuran-5-yl)-1,3,4-oxadiazole (MMBO), which displays high selectivity for GSK-3 and brain penetration following oral administration. MMBO inhibited tau phosphorylation in primary neural cell culture and also in normal mouse brain. When administered to a transgenic mouse model of AD, MMBO significantly decreased hippocampal tau phosphorylation at GSK-3 sites. Additionally, chronic MMBO administration suppressed tau pathology as assessed by AT8-immunoreactivity without affecting amyloid Ɵ pathology. Finally, in behavioral assessments, MMBO significantly improved memory and cognitive deficits in the Y-maze and in novel object recognition tests in the transgenic AD mouse model. These results indicate that pharmacological GSK-3 inhibition ameliorates behavioral dysfunction with suppression of tau phosphorylation in an AD mouse model, and that MMBO might be beneficial for AD treatment.


Subject(s)
Cognition Disorders/drug therapy , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , tau Proteins/metabolism , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Benzofurans/pharmacology , Benzofurans/therapeutic use , Brain/drug effects , Brain/metabolism , Cell Culture Techniques , Cerebral Cortex/cytology , Cognition Disorders/etiology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Peptide Fragments/metabolism , Phosphorylation/drug effects , Presenilin-1/genetics , Time Factors , tau Proteins/genetics
6.
Behav Brain Res ; 376: 112192, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31521738

ABSTRACT

Changes in the cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) signaling are implicated in older people with dementia. Drugs that modulate the cAMP/cGMP levels in the brain might therefore provide new therapeutic options for the treatment of cognitive impairment in aging and elderly with dementia. Phosphodiesterase 2A (PDE2A), which is highly expressed in the forebrain, is one of the key phosphodiesterase enzymes that hydrolyze cAMP and cGMP. In this study, we investigated the effects of PDE2A inhibition on the cognitive functions associated with aging, such as spatial learning, episodic memory, and attention, in rats with a selective, brain penetrant PDE2A inhibitor, N-{(1S)-1-[3-fluoro-4-(trifluoromethoxy)phenyl]-2-methoxyethyl-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915). Repeated treatment with TAK-915 (3Ć¢Ā€ĀÆmg/kg/day, p.o. for 4 days) significantly reduced escape latency in aged rats in the Morris water maze task compared to the vehicle treatment. In the novel object recognition task, TAK-915 (1, 3, and 10Ć¢Ā€ĀÆmg/kg, p.o.) dose-dependently attenuated the non-selective muscarinic antagonist scopolamine-induced memory deficits in rats. In addition, oral administration of TAK-915 at 10Ć¢Ā€ĀÆmg/kg significantly improved the attentional performance in middle-aged, poorly performing rats in the 5-choice serial reaction time task. These findings suggest that PDE2A inhibition in the brain has the potential to ameliorate the age-related cognitive decline.


Subject(s)
Cognitive Dysfunction/drug therapy , Pyrazines/pharmacology , Pyridines/pharmacology , Aging/drug effects , Aging/physiology , Animals , Brain/metabolism , Cognition/drug effects , Cognition Disorders/drug therapy , Cognitive Dysfunction/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Male , Memory Disorders/drug therapy , Memory, Episodic , Phosphodiesterase Inhibitors/pharmacology , Pyrazines/metabolism , Pyridines/metabolism , Rats , Rats, Inbred F344 , Rats, Long-Evans , Rats, Sprague-Dawley
7.
J Med Chem ; 60(18): 7658-7676, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28759228

ABSTRACT

Herein, we describe the discovery of a potent, selective, brain-penetrating, in vivo active phosphodiesterase (PDE) 2A inhibitor lead series. To identify high-quality leads suitable for optimization and enable validation of the physiological function of PDE2A in vivo, structural modifications of the high-throughput screening hit 18 were performed. Our lead generation efforts revealed three key potency-enhancing functionalities with minimal increases in molecular weight (MW) and no change in topological polar surface area (TPSA). Combining these structural elements led to the identification of 6-methyl-N-((1R)-1-(4-(trifluoromethoxy)phenyl)propyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (38a), a molecule with the desired balance of preclinical properties. Further characterization by cocrystal structure analysis of 38a bound to PDE2A uncovered a unique binding mode and provided insights into its observed potency and PDE selectivity. Compound 38a significantly elevated 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brain following oral administration, thus validating this compound as a useful pharmacological tool and an attractive lead for future optimization.


Subject(s)
Brain/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Administration, Oral , Animals , Brain/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Drug Discovery , Humans , Male , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats
8.
J Med Chem ; 60(18): 7677-7702, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28796496

ABSTRACT

Phosphodiesterase (PDE) 2A inhibitors have emerged as a novel mechanism with potential therapeutic option to ameliorate cognitive dysfunction in schizophrenia or Alzheimer's disease through upregulation of cyclic nucleotides in the brain and thereby achieve potentiation of cyclic nucleotide signaling pathways. This article details the expedited optimization of our recently disclosed pyrazolo[1,5-a]pyrimidine lead compound 4b, leading to the discovery of clinical candidate 36 (TAK-915), which demonstrates an appropriate combination of potency, PDE selectivity, and favorable pharmacokinetic (PK) properties, including brain penetration. Successful identification of 36 was realized through application of structure-based drug design (SBDD) to further improve potency and PDE selectivity, coupled with prospective design focused on physicochemical properties to deliver brain penetration. Oral administration of 36 demonstrated significant elevation of 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brains and improved cognitive performance in a novel object recognition task in rats. Consequently, compound 36 was advanced into human clinical trials.


Subject(s)
Brain/drug effects , Cognition/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacokinetics , Pyrazines/pharmacology , Pyrazines/pharmacokinetics , Animals , Brain/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Crystallography, X-Ray , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Drug Design , Halogenation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Phosphodiesterase Inhibitors/chemistry , Pyrazines/chemistry , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley
9.
Neurosci Res ; 80: 76-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24406748

ABSTRACT

Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) are neurodegenerative "tauopathies" characterized by hyperphosphorylated tau accumulation and neurofibrillary tangles. The P301S mutation of tau, a causal mutation of a familial type of FTLD, is believed to be involved in neurodegenerative progression. We developed a transgenic mouse, named TPR50, harboring human P301S tau. Tau phosphorylation in the hippocampus of TPR50 mice increased with age, particularly at S202/T205. Insolubilization and intracellular accumulation of tau were detected in the hippocampus by 9 months of age. Expression of calbindin was significantly reduced in 6- and 9-month-old TPR50 mice but not in 3-month-old mice. TPR50 mice demonstrated cognitive dysfunction at 5 months. At this age or earlier, although no intracellular tau accumulation was observed in the hippocampus, abnormally increased microtubule (MT)-related proteins and MT hyperdynamics in the hippocampus, and impaired axonal transport in the septo-hippocampal pathway were already observed. Therefore, cognitive dysfunction in TPR50 mice may result from early MT dysfunction and impaired axonal transport rather than accumulation of insoluble tau and neurodegeneration. TPR50 mice are a valuable new model to study progression of tauopathies at both the behavioral and neurocellular levels and may also prove useful for testing new therapies for neurodegenerative diseases.


Subject(s)
Axonal Transport/genetics , Cognition Disorders/genetics , Mutation/genetics , Proline/genetics , Serine/genetics , tau Proteins/genetics , Age Factors , Animals , Cognition Disorders/pathology , Disease Progression , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Humans , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Recognition, Psychology/physiology , tau Proteins/metabolism
10.
Brain Res ; 1296: 148-63, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19698704

ABSTRACT

Neurofibrillary tangles (NFTs) composed of hyperphosphorylated and aggregated tau are common pathological characteristics in Alzheimer's disease (AD) and other tauopathies. Aberrant tau phosphorylation is an early and pivotal event in the pathogenesis of tauopathies, and since GSK-3 is a key factor implicated in aberrant tau phosphorylation, GSK-3 inhibition is expected to suppress tauopathy disease progression. In the present study, we report the efficacy of a newly discovered small molecule GSK-3 inhibitor, 6-methyl-N-[3-[[3-(1-methylethoxy)propyl]carbamoyl]-1H-pyrazol-4-yl]pyridine-3-carboxamide (compound A), to inhibit tau phosphorylation and to reduce the amount of pathological aggregated tau in JNPL3 mice that overexpress a mutant form of human tau. Compound A is a highly potent and selective inhibitor of GSK-3 with an IC(50) of 2 nM, with at least 230-fold lower potency against 27 other kinases. Oral administration of compound A resulted in a significant reduction of tau phosphorylation at several GSK-3 directed sites. Furthermore, chronic oral administration of compound A markedly reduced aggregated tau in old JNPL3 mice. These results suggest that a novel, orally active GSK-3 inhibitor, compound A, has potency in the prevention of tau pathology.


Subject(s)
Brain/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Niacinamide/analogs & derivatives , Pyrazoles/pharmacology , tau Proteins/metabolism , Administration, Oral , Aging , Animals , Brain/metabolism , Cold Temperature , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Niacinamide/administration & dosage , Niacinamide/chemistry , Niacinamide/pharmacology , Phosphorylation/drug effects , Protein Multimerization/drug effects , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Stress, Physiological/drug effects , Time Factors , tau Proteins/genetics
11.
J Med Chem ; 52(20): 6270-86, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19775160

ABSTRACT

Glycogen synthase kinase 3beta (GSK-3beta) inhibition is expected to be a promising therapeutic approach for treating Alzheimer's disease. Previously we reported a series of 1,3,4-oxadiazole derivatives as potent and highly selective GSK-3beta inhibitors, however, the representative compounds 1a,b showed poor pharmacokinetic profiles. Efforts were made to address this issue by reducing molecular weight and lipophilicity, leading to the identification of oxadiazole derivatives containing a sulfinyl group, (S)-9b and (S)-9c. These compounds exhibited not only highly selective and potent inhibitory activity against GSK-3beta but also showed good pharmacokinetic profiles including favorable BBB penetration. In addition, (S)-9b and (S)-9c given orally to mice significantly inhibited cold water stress-induced tau hyperphosphorylation in mouse brain.


Subject(s)
Brain/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Oxadiazoles/metabolism , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Crystallography, X-Ray , Drug Design , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3 beta , Humans , Inhibitory Concentration 50 , Male , Mice , Models, Molecular , Molecular Conformation , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Permeability , Protein Kinase Inhibitors/analogs & derivatives , Protein Kinase Inhibitors/pharmacokinetics , Rats , Solubility , Stereoisomerism , Substrate Specificity
12.
Am J Respir Cell Mol Biol ; 35(1): 103-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16528013

ABSTRACT

Airway hyperresponsiveness (AHR) is an important feature of bronchial asthma. Although the incidence of AHR has genetic and environmental components, the mechanism of AHR in asthma remains unclear. The identification of genes that are preferentially expressed in a murine model of AHR could help elucidate the molecular mechanisms of this pulmonary pathology. Suppressive subtractive hybridization analysis revealed that eosinophil chemotactic factor by T lymphocytes (ECF-L), a mouse chitinase family protein, was selectively expressed in the lungs of mice with AHR. Induction of ECF-L expression was observed soon after allergen exposure but before the onset of airway inflammation. Cell-specific ECF-L expression was examined by in situ hybridization using digoxigenin-labeled antisense RNA probes and immunofluorescence staining. The assay revealed that the ECF-L-expressing cells in the lungs of the AHR-model mice are alveolar macrophages. Intratracheal administration of an adenoviral vector that expressed antisense ECF-L RNA (Ad-ECF-L-AS) suppressed AHR and eosinophil infiltration. These results indicate that ECF-L may play a critical role in allergic inflammation and bronchial asthma.


Subject(s)
Asthma/metabolism , Bronchial Hyperreactivity/metabolism , Chemokines/metabolism , Chemotactic Factors, Eosinophil/metabolism , Disease Models, Animal , T-Lymphocytes/metabolism , Adenoviridae/genetics , Animals , Asthma/immunology , Bronchial Hyperreactivity/immunology , Chemokines/genetics , Chemotactic Factors, Eosinophil/genetics , Gene Expression Profiling , Lung/cytology , Lung/pathology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred BALB C , RNA, Antisense/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trachea/metabolism
13.
Am J Respir Crit Care Med ; 165(8): 1132-6, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11956057

ABSTRACT

Mucus overproduction is a clinical feature of asthma. Ca2+-activated Cl- channel 1 (CaCC1) has been identified as a protein that is expressed in intestinal epithelia and that plays an important role in fluid and electrolyte transport. Recently, its mouse counterpart, gob-5, was identified as a key molecule in the induction of murine asthma through mucus overproduction. To elucidate the relationship of CaCC1 to human asthma, we examined CaCC1 expression using real-time quantitative polymerase chain reaction analysis in bronchial tissues from patients with asthma and normal control subjects. The expression of CaCC1 was significantly upregulated in patients with bronchial asthma compared with control subjects. In situ hybridization and immunohistochemical analysis demonstrated that CaCC1 is located in the bronchial epithelium, especially in mucus-producing goblet cells. In vitro transfection of a CaCC1 expression vector into the human mucoepidermoid cell line, NCI-H292, increased mucus production and induced the MUC5AC gene. These results suggest that CaCC1 plays a direct role in mucus production and differentiation in goblet cells and may contribute to the pathogenesis of asthma through its mucus-inducing activity.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Calcium/metabolism , Chloride Channels/metabolism , Gene Expression Regulation , Asthma/genetics , Asthma/physiopathology , Bronchi/cytology , Cell Line , Chloride Channels/genetics , Goblet Cells/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Mucin 5AC , Mucins , Mucus/metabolism , Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL