Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Neurosci ; 102: 103449, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31770590

RESUMEN

A novel, potent, and highly specific inhibitor of calcium-calmodulin-dependent phosphodiesterases (PDE) of the PDE1 family, ITI-214, was used to investigate the role of PDE1 in inflammatory responses. ITI-214 dose-dependently suppressed lipopolysaccharide (LPS)-induced gene expression of pro-inflammatory cytokines in an immortalized murine microglial cell line, BV2 cells. RNA profiling (RNA-Seq) was used to analyze the impact of ITI-214 on the BV2 cell transcriptome in the absence and the presence of LPS. ITI-214 was found to regulate classes of genes that are involved in inflammation and cell migration responses to LPS exposure. The gene expression changes seen with ITI-214 treatment were distinct from those elicited by inhibitors of other PDEs with anti-inflammatory activity (e.g., a PDE4 inhibitor), indicating a distinct mechanism of action for PDE1. Functionally, ITI-214 inhibited ADP-induced migration of BV2 cells through a P2Y12-receptor-dependent pathway, possibly due to increases in the extent of cAMP and VASP phosphorylation downstream of receptor activation. Importantly, this effect was recapitulated in P2 rat microglial cells in vitro, indicating that these pathways are active in native microglial cells. These studies are the first to demonstrate that inhibition of PDE1 exerts anti-inflammatory effects through effects on microglia signaling pathways. The ability of PDE1 inhibitors to prevent or dampen excessive inflammatory responses of BV2 cells and microglia provides a basis for exploring their therapeutic utility in the treatment of neurodegenerative diseases associated with increased inflammation and microglia proliferation such as Parkinson's disease and Alzheimer's disease.


Asunto(s)
Antiinflamatorios/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Microglía/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Movimiento Celular , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/fisiología , Fosfoproteínas/metabolismo , Ratas , Receptores Purinérgicos P2Y12/metabolismo , Transducción de Señal
2.
Cell Signal ; 62: 109338, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31176021

RESUMEN

α7 nicotinic acetylcholine receptors (α7nAChRs) have been targeted to improve cognition in different neurological and psychiatric disorders. Nevertheless, no α7nAChR activating ligand has been clinically approved. Here, we investigated the effects of antagonizing α7nAChRs using the selective antagonist methyllycaconitine (MLA) on receptor activity in vitro and cognitive functioning in vivo. Picomolar concentrations of MLA significantly potentiated receptor responses in electrophysiological experiments mimicking the in vivo situation. Furthermore, microdialysis studies showed that MLA administration substantially increased hippocampal glutamate efflux which is related to memory processes. Accordingly, pre-tetanus administration of low MLA concentrations produced longer lasting potentiation (long-term potentiation, LTP) in studies examining hippocampal plasticity. Moreover, low doses of MLA improved acquisition, but not consolidation memory processes in rats. While the focus to enhance cognition by modulating α7nAChRs lies on agonists and positive modulators, antagonists at low doses should provide a novel approach to improve cognition in neurological and psychiatric disorders.


Asunto(s)
Aconitina/análogos & derivados , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/genética , Aconitina/metabolismo , Aconitina/farmacología , Animales , Cognición/fisiología , Modelos Animales de Enfermedad , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Memoria/fisiología , Antagonistas Nicotínicos/farmacología , Ratas , Receptores Nicotínicos/genética , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
3.
Circulation ; 138(18): 1974-1987, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30030415

RESUMEN

BACKGROUND: Phosphodiesterase type-1 (PDE1) hydrolyzes cAMP and cGMP and is constitutively expressed in the heart, although cardiac effects from its acute inhibition in vivo are largely unknown. Existing data are limited to rodents expressing mostly the cGMP-favoring PDE1A isoform. Human heart predominantly expresses PDE1C with balanced selectivity for cAMP and cGMP. Here, we determined the acute effects of PDE1 inhibition in PDE1C-expressing mammals, dogs, and rabbits, in normal and failing hearts, and explored its regulatory pathways. METHODS: Conscious dogs chronically instrumented for pressure-volume relations were studied before and after tachypacing-induced heart failure (HF). A selective PDE1 inhibitor (ITI-214) was administered orally or intravenously±dobutamine. Pressure-volume analysis in anesthetized rabbits tested the role of ß-adrenergic and adenosine receptor signaling on ITI-214 effects. Sarcomere and calcium dynamics were studied in rabbit left ventricular myocytes. RESULTS: In normal and HF dogs, ITI-214 increased load-independent contractility, improved relaxation, and reduced systemic arterial resistance, raising cardiac output without altering systolic blood pressure. Heart rate increased, but less so in HF dogs. ITI-214 effects were additive to ß-adrenergic receptor agonism (dobutamine). Dobutamine but not ITI-214 increased plasma cAMP. ITI-214 induced similar cardiovascular effects in rabbits, whereas mice displayed only mild vasodilation and no contractility effects. In rabbits, ß-adrenergic receptor blockade (esmolol) prevented ITI-214-mediated chronotropy, but inotropy and vasodilation remained unchanged. By contrast, adenosine A2B-receptor blockade (MRS-1754) suppressed ITI-214 cardiovascular effects. Adding fixed-rate atrial pacing did not alter the findings. ITI-214 alone did not affect sarcomere or whole-cell calcium dynamics, whereas ß-adrenergic receptor agonism (isoproterenol) or PDE3 inhibition (cilostamide) increased both. Unlike cilostamide, which further enhanced shortening and peak calcium when combined with isoproterenol, ITI-214 had no impact on these responses. Both PDE1 and PDE3 inhibitors increased shortening and accelerated calcium decay when combined with forskolin, yet only cilostamide increased calcium transients. CONCLUSIONS: PDE1 inhibition by ITI-214 in vivo confers acute inotropic, lusitropic, and arterial vasodilatory effects in PDE1C-expressing mammals with and without HF. The effects appear related to cAMP signaling that is different from that provided via ß-adrenergic receptors or PDE3 modulation. ITI-214, which has completed phase I trials, may provide a novel therapy for HF.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Miocitos Cardíacos/fisiología , Animales , Calcio/metabolismo , AMP Cíclico/sangre , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Dobutamina/uso terapéutico , Perros , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Conejos , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Mol Cell Neurosci ; 88: 292-299, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29501719

RESUMEN

Stress during pregnancy increases the risk to develop psychological disorders such as depression during pregnancy or in the postpartum period. According to the neurotrophin hypothesis of depression, the pathophysiology of depression is caused by reduced neurotrophic activity in the brain. However, most studies only focus on the molecular changes happening to the offspring upon gestational stress. To gain insight into the potential molecular changes happening in the stressed dams, C57Bl6/J mice were stressed during their first week of gestation. At 28 days postpartum, the hippocampus and nucleus accumbens core of the dams, two brain regions heavily implicated in depression, were evaluated using immunohistochemistry to detect changes in the neurotrophin system. Gestational stress decreased the weight of the dams, increased the chance for spontaneous abortion and increased the weight of offspring. Litter size, survival rates and sex distribution were not altered as a consequence of gestational stress. Hippocampal brain-derived neurotrophic factor (BDNF) decreased following exposure to stress during pregnancy. Hippocampal protein levels of p75NTR, a low-affinity receptor for BDNF which can induce apoptosis, were increased following exposure to stress. Protein levels of p11, of which the expression is regulated by BDNF, were decreased in the hippocampus. No changes were found for TrkB immunostaining or apoptosis. Taken together, this shows that stress during pregnancy negatively affects the neurotrophin system in the hippocampus of the dams, thereby reducing hippocampal plasticity. These data confirm that gestational stress has a negative impact on pregnancy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Péptidos de Penetración Celular/metabolismo , Hipocampo/metabolismo , Periodo Posparto/psicología , Animales , Apoptosis/fisiología , Conducta Animal , Corticosterona/metabolismo , Femenino , Ratones Endogámicos C57BL , Embarazo , Estrés Psicológico/fisiopatología
5.
Adv Neurobiol ; 17: 349-384, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28956339

RESUMEN

The focus of this chapter is on the cyclic nucleotide phosphodiesterase 1 (PDE1) family. PDE1 is one member of the 11 PDE families (PDE 1-11). It is the only phosphodiesterase family that is calcium/calmodulin activated. As a result, whereas other families of PDEs 2-11 play a dominant role controlling basal levels of cyclic nucleotides, PDE1 is involved when intra-cellular calcium levels are elevated and, thus, has an "on demand" or activity-dependent involvement in the control of cyclic nucleotides in excitatory cells including neurons, cardiomyocytes and smooth muscle. As a Class 1 phosphodiesterase, PDE1 hydrolyzes the 3' bond of 3'-5'-cyclic nucleotides, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Here, we review evidence for this family of enzymes as drug targets for development of therapies aimed to address disorders of the central nervous system (CNS) and of degenerative diseases. The chapter includes sections on the potential for cognitive enhancement in mental disorders, as well as a review of PDE1 enzyme structure, enzymology, tissue distribution, genomics, inhibitors, pharmacology, clinical trials, and therapeutic indications. Information is taken from public databases. A number of excellent reviews of the phosphodiesterase family have been written as well as reviews of the PDE1 family. References cited here are not comprehensive, rather pointing to major reviews and key publications.


Asunto(s)
Disfunción Cognitiva/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Trastornos Mentales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Humanos , Trastornos Mentales/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico
6.
Psychopharmacology (Berl) ; 233(17): 3113-24, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27342643

RESUMEN

RATIONALE: Therapeutic agents for memory enhancement in psychiatric disorders, such as schizophrenia, are urgently needed. OBJECTIVE: The aim of this study is to characterize the preclinical profile of ITI-214, a potent inhibitor of phosphodiesterase 1 (PDE1). METHODS: ITI-214 was assayed for inhibition of PDE1 versus other PDE enzyme families using recombinant human PDE enzymes and for off-target binding to 70 substrates (General SEP II diversity panel; Caliper Life Sciences). Effects of ITI-214 (0.1-10 mg/kg, po) on memory performance were assayed in rats using the novel object recognition (NOR) paradigm, with drug given at specified time points prior to or following exposure to objects in an open field. ITI-214 was evaluated for potential drug-drug interaction with risperidone in rats using conditioned avoidance response (CAR) and pharmacokinetic assessments. RESULTS: ITI-214 inhibited PDE1A (K i = 33 pmol) with >1000-fold selectivity for the nearest other PDE family (PDE4D) and displayed minimal off-target binding interactions in a 70-substrate selectivity profile. By using specific timing of oral ITI-214 administration, it was demonstrated in the NOR that ITI-214 is able to enhance acquisition, consolidation, and retrieval memory processes. All memory effects were in the absence of effects on exploratory behavior. ITI-214 did not disrupt the risperidone pharmacokinetic profile or effects in CAR. CONCLUSIONS: ITI-214 improved the memory processes of acquisition, consolidation, and retrieval across a broad dose range (0.1-10 mg/kg, po) without disrupting the antipsychotic-like activity of a clinical antipsychotic medication, specifically risperidone. Clinical development of ITI-214 is currently in progress.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Memoria/efectos de los fármacos , Nootrópicos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Animales , Antipsicóticos/farmacología , Interacciones Farmacológicas , Conducta Exploratoria/efectos de los fármacos , Masculino , Ratas , Risperidona/farmacología , Esquizofrenia , Psicología del Esquizofrénico
7.
Int J Neuropsychopharmacol ; 19(10)2016 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-27037577

RESUMEN

BACKGROUND: The fronto-striatal circuits are the common neurobiological basis for neuropsychiatric disorders, including schizophrenia, Parkinson's disease, Huntington's disease, attention deficit hyperactivity disorder, obsessive-compulsive disorder, and Tourette's syndrome. Fronto-striatal circuits consist of motor circuits, associative circuits, and limbic circuits. All circuits share 2 common features. First, all fronto-striatal circuits consist of hyper direct, direct, and indirect pathways. Second, all fronto-striatal circuits are modulated by dopamine. Intracellularly, the effect of dopamine is largely mediated through the cyclic adenosine monophosphate/protein kinase A signaling cascade with an additional role for the cyclic guanosine monophosphate/protein kinase G pathway, both of which can be regulated by phosphodiesterases. Phosphodiesterases are thus a potential target for pharmacological intervention in neuropsychiatric disorders related to dopaminergic regulation of fronto-striatal circuits. METHODS: Clinical studies of the effects of different phosphodiesterase inhibitors on cognition, affect, and motor function in relation to the fronto-striatal circuits are reviewed. RESULTS: Several selective phosphodiesterase inhibitors have positive effects on cognition, affect, and motor function in relation to the fronto-striatal circuits. CONCLUSION: Increased understanding of the subcellular localization and unraveling of the signalosome concept of phosphodiesterases including its function and dysfunction in the fronto-striatal circuits will contribute to the design of new specific inhibitors and enhance the potential of phosphodiesterase inhibitors as therapeutics in fronto-striatal circuits.

8.
J Med Chem ; 59(3): 1149-64, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26789933

RESUMEN

A diverse set of 3-aminopyrazolo[3,4-d]pyrimidinones was designed and synthesized. The structure-activity relationships of these polycyclic compounds as phosphodiesterase 1 (PDE1) inhibitors were studied along with their physicochemical and pharmacokinetic properties. Systematic optimizations of this novel scaffold culminated in the identification of a clinical candidate, (6aR,9aS)-2-(4-(6-fluoropyridin-2-yl)benzyl)-5-methyl-3-(phenylamino)-5,6a,7,8,9,9a-hexahydrocyclopenta[4,5]imidazo[1,2-a]pyrazolo[4,3-e]pyrimidin-4-(2H)-one phosphate (ITI-214), which exhibited picomolar inhibitory potency for PDE1, demonstrated excellent selectivity against all other PDE families and showed good efficacy in vivo. Currently, this investigational new drug is in Phase I clinical development and being considered for the treatment of several indications including cognitive deficits associated with schizophrenia and Alzheimer's disease, movement disorders, attention deficit and hyperactivity disorders, and other central nervous system (CNS) and non-CNS disorders.


Asunto(s)
Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Descubrimiento de Drogas , Trastornos Mentales/complicaciones , Enfermedades Neurodegenerativas/complicaciones , Inhibidores de Fosfodiesterasa/farmacología , Animales , Bovinos , Trastornos del Conocimiento/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/enzimología , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Estructura Molecular , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/metabolismo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
9.
Psychopharmacology (Berl) ; 232(3): 605-21, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25120104

RESUMEN

RATIONALE: Schizophrenia remains among the most prevalent neuropsychiatric disorders, and current treatment options are accompanied by unwanted side effects. New treatments that better address core features of the disease with minimal side effects are needed. OBJECTIVES: As a new therapeutic approach, 1-(4-fluoro-phenyl)-4-((6bR, 10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H,7H-pyrido[3',4':4,5]pyrrolo[1,2,3-de]quinoxalin-8-yl)-butan-1-one (ITI-007) is currently in human clinical trials for the treatment of schizophrenia. Here, we characterize the preclinical functional activity of ITI-007. RESULTS: ITI-007 is a potent 5-HT2A receptor ligand (K i = 0.5 nM) with strong affinity for dopamine (DA) D2 receptors (K i = 32 nM) and the serotonin transporter (SERT) (K i = 62 nM) but negligible binding to receptors (e.g., H1 histaminergic, 5-HT2C, and muscarinic) associated with cognitive and metabolic side effects of antipsychotic drugs. In vivo it is a 5-HT2A antagonist, blocking (±)-2,5-dimethoxy-4-iodoamphetamine hydrochloride (DOI)-induced headtwitch in mice with an inhibitory dose 50 (ID50) = 0.09 mg/kg, per oral (p.o.), and has dual properties at D2 receptors, acting as a postsynaptic D2 receptor antagonist to block D-amphetamine hydrochloride (D-AMPH) hyperlocomotion (ID50 = 0.95 mg/kg, p.o.), yet acting as a partial agonist at presynaptic striatal D2 receptors in assays measuring striatal DA neurotransmission. Further, in microdialysis studies, this compound significantly and preferentially enhances mesocortical DA release. At doses relevant for antipsychotic activity in rodents, ITI-007 has no demonstrable cataleptogenic activity. ITI-007 indirectly modulates glutamatergic neurotransmission by increasing phosphorylation of GluN2B-type N-methyl-D-aspartate (NMDA) receptors and preferentially increases phosphorylation of glycogen synthase kinase 3ß (GSK-3ß) in mesolimbic/mesocortical dopamine systems. CONCLUSION: The combination of in vitro and in vivo activities of this compound support its development for the treatment of schizophrenia and other psychiatric and neurologic disorders.


Asunto(s)
Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Ácido Glutámico/metabolismo , Neurotransmisores/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Descubrimiento de Drogas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
10.
J Med Chem ; 57(6): 2670-82, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24559051

RESUMEN

We report the synthesis and structure-activity relationships of a class of tetracyclic butyrophenones that exhibit potent binding affinities to serotonin 5-HT(2A) and dopamine D2 receptors. This work has led to the discovery of 4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H,7H-pyrido[3',4':4,5]pyrrolo[1,2,3-de]quinoxalin-8-yl)-1-(4-fluorophenyl)-butan-1-one 4-methylbenzenesulfonate (ITI-007), which is a potent 5-HT(2A) antagonist, postsynaptic D2 antagonist, and inhibitor of serotonin transporter. This multifunctional drug candidate is orally bioavailable and exhibits good antipsychotic efficacy in vivo. Currently, this investigational new drug is under clinical development for the treatment of neuropsychiatric and neurological disorders.


Asunto(s)
Trastornos Mentales/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Quinoxalinas/síntesis química , Quinoxalinas/farmacología , Inhibidores de Captación Adrenérgica/síntesis química , Inhibidores de Captación Adrenérgica/farmacología , Animales , Conducta Animal/efectos de los fármacos , Disponibilidad Biológica , Descubrimiento de Drogas , Electrochoque , Indicadores y Reactivos , Masculino , Quinoxalinas/farmacocinética , Quipazina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/química , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/metabolismo , Receptores de Dopamina D2/química , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Proteínas Recombinantes/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Antagonistas de la Serotonina/síntesis química , Antagonistas de la Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Relación Estructura-Actividad
11.
Nature ; 467(7311): 95-8, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20811458

RESUMEN

Accumulation of neurotoxic amyloid-beta is a major hallmark of Alzheimer's disease. Formation of amyloid-beta is catalysed by gamma-secretase, a protease with numerous substrates. Little is known about the molecular mechanisms that confer substrate specificity on this potentially promiscuous enzyme. Knowledge of the mechanisms underlying its selectivity is critical for the development of clinically effective gamma-secretase inhibitors that can reduce amyloid-beta formation without impairing cleavage of other gamma-secretase substrates, especially Notch, which is essential for normal biological functions. Here we report the discovery of a novel gamma-secretase activating protein (GSAP) that drastically and selectively increases amyloid-beta production through a mechanism involving its interactions with both gamma-secretase and its substrate, the amyloid precursor protein carboxy-terminal fragment (APP-CTF). GSAP does not interact with Notch, nor does it affect its cleavage. Recombinant GSAP stimulates amyloid-beta production in vitro. Reducing GSAP concentrations in cell lines decreases amyloid-beta concentrations. Knockdown of GSAP in a mouse model of Alzheimer's disease reduces levels of amyloid-beta and plaque development. GSAP represents a type of gamma-secretase regulator that directs enzyme specificity by interacting with a specific substrate. We demonstrate that imatinib, an anticancer drug previously found to inhibit amyloid-beta formation without affecting Notch cleavage, achieves its amyloid-beta-lowering effect by preventing GSAP interaction with the gamma-secretase substrate, APP-CTF. Thus, GSAP can serve as an amyloid-beta-lowering therapeutic target without affecting other key functions of gamma-secretase.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Benzamidas , Línea Celular , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Mesilato de Imatinib , Ratones , Fragmentos de Péptidos/metabolismo , Piperazinas/farmacología , Proteínas/genética , Pirimidinas/farmacología , Interferencia de ARN , Receptor Notch1/metabolismo
12.
Bioorg Med Chem Lett ; 20(16): 4825-30, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20638843

RESUMEN

Two distinct families of small molecules were discovered as novel alpha7 nicotinic acetylcholine receptor (nAChR) antagonists by pharmacophore-based virtual screening. These novel antagonists exhibited selectivity for the neuronal alpha7 subtype over other nAChRs and good brain penetration. Neuroprotection was demonstrated by representative compounds 7i and 8 in a mouse seizure-like behavior model induced by the nerve agent diisopropylfluorophosphate (DFP). These novel nAChR antagonists have potential use as antidote for organophosphorus nerve agent intoxication.


Asunto(s)
Antagonistas Nicotínicos/química , Receptores Nicotínicos/química , Animales , Encéfalo/metabolismo , Simulación por Computador , Modelos Animales de Enfermedad , Humanos , Ratones , Conformación Molecular , Antagonistas Nicotínicos/síntesis química , Antagonistas Nicotínicos/uso terapéutico , Unión Proteica , Ratas , Receptores Nicotínicos/metabolismo , Convulsiones/tratamiento farmacológico , Relación Estructura-Actividad , Receptor Nicotínico de Acetilcolina alfa 7
13.
Brain Res ; 1342: 11-23, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20423708

RESUMEN

Organophosphorus (OP) compounds cause toxic symptoms, including convulsions, coma, and death, as the result of irreversible inhibition of acetylcholinesterase (AChE). The development of effective treatments to block these effects and attenuate long-term cognitive and motor disabilities that result from OP intoxication is hampered by a limited understanding of the CNS pathways responsible for these actions. We employed a candidate method (called CNSProfile) to identify changes in the phosphorylation state of key neuronal phosphoproteins evoked by the OP compound, diisopropyl fluorophosphate (DFP). Focused microwave fixation was used to preserve the phosphorylation state of phosphoproteins in brains of DFP-treated mice; hippocampus and striatum were analyzed by immunoblotting with a panel of phospho-specific antibodies. DFP exposure elicited comparable effects on phosphorylation of brain phosphoproteins in both C57BL/6 and FVB mice. DFP treatment significantly altered phosphorylation at regulatory residues on glutamate receptors, including Serine897 (S897) of the NR1 NMDA receptor. NR1 phosphorylation was bi-directionally regulated after DFP in striatum versus hippocampus. NR1 phosphorylation was reduced in striatum, but elevated in hippocampus, compared with controls. DARPP-32 phosphorylation in striatum was selectively increased at the Cdk5 kinase substrate, Threonine75 (T75). Phencynonate hydrochloride, a muscarinic cholinergic antagonist, prevented seizure-like behaviors and the observed changes in phosphorylation induced by DFP. The data reveal region-specific effects of nerve agent exposure on intracellular signaling pathways that correlate with seizure-like behavior and which are reversed by the muscarinic receptor blockade. This approach identifies specific targets for nerve agents, including substrates for Cdk5 kinase, which may be the basis for new anti-convulsant therapies.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/toxicidad , Isoflurofato/toxicidad , Proteínas del Tejido Nervioso/efectos de los fármacos , Animales , Encéfalo/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Microondas , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
14.
J Nat Prod ; 72(12): 2199-202, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19994845

RESUMEN

The bis-indole alkaloid indirubin and its analogues bear a very interesting natural pharmacophore. They are recognized mainly as kinase inhibitors, but several other activities make them possible candidates for preclinical studies. Based on the previously reported activity of 7-bromoindirubin-3'-oxime and its derivatives, the synthesis of indirubins bearing a heterocyclic nitrogen atom at position 7 was carried out. Herein, we report the first synthesis of 7-azaindirubin-3'-oxime (12) as well as its antiproliferative activity against 57 cancer cell lines and its inhibitory activity against a series of kinases. 7-Azaindirubin (10) and its 3'-oxime derivative (12) showed reduced activity as kinase inhibitors in comparison with other known indirubin derivatives, but antiproliferative activity with a best GI(50) value of 0.77 microM.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/síntesis química , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/farmacología , Alcaloides Indólicos/síntesis química , Alcaloides Indólicos/farmacología , Secuencia de Aminoácidos , Antineoplásicos/química , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Alcaloides Indólicos/química , Indoles/farmacología , Estructura Molecular
15.
J Biol Chem ; 282(5): 2911-7, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17135270

RESUMEN

Protein tyrosine phosphatase 1B (PTP-1B) has been implicated in the regulation of the insulin receptor. Dephosphorylation of the insulin receptor results in decreased insulin signaling and thus decreased glucose uptake. PTP-1B-/- mice have increased insulin sensitivity and are resistant to weight gain when fed a high fat diet, validating PTP-1B as a potential target for the treatment of type 2 diabetes. Many groups throughout the world have been searching for selective inhibitors for PTP-1B, and most of them target inhibitors to PTP-1B-(1-298), the N-terminal catalytic domain of the enzyme. However, the C-terminal domain is quite large and could influence the activity of the enzyme. Using two constructs of PTP-1B and a phosphopeptide as substrate, steady state assays showed that the presence of the C-terminal domain decreased both the Km and the k(cat) 2-fold. Pre-steady state kinetic experiments showed that the presence of the C-terminal domain improved the affinity of the enzyme for a phosphopeptide 2-fold, primarily because the off-rate was slower. This suggests that the C-terminal domain of PTP-1B may contact the phosphopeptide in some manner, allowing it to remain at the active site longer. This could be useful when screening libraries of compounds for inhibitors of PTP-1B. A compound that is able to make contacts with the C-terminal domain of PTP-1B would not only have a modest improvement in affinity but may also provide for specificity over other phosphatases.


Asunto(s)
Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cartilla de ADN , Humanos , Cinética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Fosfatasas/genética , Proteínas Recombinantes/metabolismo
16.
FEBS Lett ; 579(2): 464-8, 2005 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-15642359

RESUMEN

The C-Raf kinase is regulated by numerous phosphorylation steps. To quantify the most prominent phosphorylation sites of C-Raf, we performed mass spectrometry analysis of wild-type C-Raf and the constitutively active C-Raf mutant C-Raf-Y340D/Y341D. We confirmed phosphorylation of most of the sites reported in the literature with the exception that we did not detect phosphorylation of threonine 268/269 (autophosphorylation sites) and threonine 491/serine 494 (kinase activation loop). Importantly, we detected novel phosphorylation sites at the positions of serine 296 and 301. The degree of phosphorylation in these positions depends on the level of activation of C-Raf. Furthermore, we show here, using point mutant forms of C-Raf kinases with serine to alanine and serine to aspartic acid substitution, that serines 296 and 301 contribute to negative regulation of C-Raf.


Asunto(s)
Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Serina/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Dominio Catalítico/genética , Chlorocebus aethiops , Humanos , Datos de Secuencia Molecular , Mutación/genética , Mapeo Peptídico , Fosforilación , Proteínas Proto-Oncogénicas c-raf/genética , Alineación de Secuencia , Serina/genética
17.
Genomics ; 81(2): 112-25, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12620389

RESUMEN

We have performed an exhaustive unbiased yeast two-hybrid analysis to identify interaction partners of two human Raf kinase isoforms, A-Raf and C-Raf, using their N-terminal regulatory domain as "bait." A total of 20 different human proteins were found to interact with Raf isoforms. Several of these interactions were novel and an extensive bioinformatics evaluation was performed for each. The novel putative interactions include a signalosome component, TOPK/PBK kinase, and two new putative protein phosphatases. The cysteine-rich zinc-binding domain (CRD) of Raf was found to interact with all 20 proteins and to achieve isoform-specific interactions. Since similar putative CRDs are present in a variety of protein serine-threonine kinases, the data suggest that the CRD may function as a major protein-protein interaction domain of these kinases. We propose possible functional consequences of these novel Raf interactions.


Asunto(s)
Proteínas Proto-Oncogénicas c-raf/metabolismo , Técnicas del Sistema de Dos Híbridos , Secuencia de Aminoácidos , Animales , Bacterias/metabolismo , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas A-raf
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...