Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(2): 211-221, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-27777420

RESUMO

Here, we report a novel target of the drug memantine, ATP-sensitive K+ (KATP) channels, potentially relevant to memory improvement. We confirmed that memantine antagonizes memory impairment in Alzheimer's model APP23 mice. Memantine increased CaMKII activity in the APP23 mouse hippocampus, and memantine-induced enhancement of hippocampal long-term potentiation (LTP) and CaMKII activity was totally abolished by treatment with pinacidil, a specific opener of KATP channels. Memantine also inhibited Kir6.1 and Kir6.2 KATP channels and elevated intracellular Ca2+ concentrations in neuro2A cells overexpressing Kir6.1 or Kir6.2. Kir6.2 was preferentially expressed at postsynaptic regions of hippocampal neurons, whereas Kir6.1 was predominant in dendrites and cell bodies of pyramidal neurons. Finally, we confirmed that Kir6.2 mutant mice exhibit severe memory deficits and impaired hippocampal LTP, impairments that cannot be rescued by memantine administration. Altogether, our studies show that memantine modulates Kir6.2 activity, and that the Kir6.2 channel is a novel target for therapeutics to improve memory impairment in Alzheimer disease patients.


Assuntos
Memantina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/efeitos dos fármacos , Dendritos , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Memantina/metabolismo , Memória/efeitos dos fármacos , Memória/fisiologia , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Neurônios , Fosforilação , Canais de Potássio/efeitos dos fármacos , Células Piramidais , Sinapses , Lobo Temporal
2.
Mol Psychiatry ; 22(8): 1205-1222, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27922607

RESUMO

Aberrant dopamine D2 receptor (D2R) activity is associated with neuropsychiatric disorders, making those receptors targets for antipsychotic drugs. Here, we report that novel signaling through the intracellularly localized D2R long isoform (D2LR) elicits extracellular signal-regulated kinase (ERK) activation and dendritic spine formation through Rabex-5/platelet-derived growth factor receptor-ß (PDGFRß)-mediated endocytosis in mouse striatum. We found that D2LR directly binds to and activates Rabex-5, promoting early-endosome formation. Endosomes containing D2LR and PDGFRß are then transported to the Golgi apparatus, where those complexes trigger Gαi3-mediated ERK signaling. Loss of intracellular D2LR-mediated ERK activation decreased neuronal activity and dendritic spine density in striatopallidal medium spiny neurons (MSNs). In addition, dendritic spine density in striatopallidal MSNs significantly increased following treatment of striatal slices from wild-type mice with quinpirole, a D2R agonist, but those changes were lacking in D2LR knockout mice. Moreover, intracellular D2LR signaling mediated effects of a typical antipsychotic drug, haloperidol, in inducing catalepsy behavior. Taken together, intracellular D2LR signaling through Rabex-5/PDGFRß is critical for ERK activation, dendritic spine formation and neuronal activity in striatopallidal MSNs of mice.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Técnicas de Cultura de Células , Corpo Estriado/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/fisiologia , Agonistas de Dopamina/farmacologia , Endocitose/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Haloperidol/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação , Isoformas de Proteínas , Quimpirol/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
3.
Neuroscience ; 234: 103-15, 2013 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-23313709

RESUMO

Neonatal ventral hippocampus (NVH)-lesioned rats represent a neurodevelopmental impairment model of schizophrenia. Previous observations indicate that postpubertal NVH-lesioned rats exhibit impairments in prepulse inhibition (PPI), spontaneous locomotion and social interaction behavior. Here, we document the neurochemical basis of those defects. PPI impairment but not cognitive impairment was improved by acute risperidone treatment (0.30mg/kgi.p.). Immunohistochemical analyses using anti-autophosphorylated Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) antibody indicated significantly reduced CaMKII autophosphorylation, especially in the medial prefrontal cortex (mPFC), striatum and hippocampal CA1 region, of NVH-lesioned rats relative to control animals. We also confirmed that reduced CaMKII autophoshorylation in the mPFC, striatum and hippocampal CA1 region causes decreased phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolpropionic acid-type glutamate receptor subunit 1 (GluR1) (Ser 831), a CaMKII substrate. Like CaMKII, PKCα (Ser 657) autophosphorylation and NR1 (Ser 896) phosphorylation were decreased both in the mPFC and CA1 region. Interestingly, phosphorylation of DARPP-32 (Thr 34) was decreased in the mPFC but increased in the striatum and CA1 region of NVH-lesioned rats compared to controls. Risperidone treatment restored increased DARPP-32 phosphorylation in the striatum and CA1 regions of NVH-lesioned rats but did not rescue CaMKII and PKCα autophosphorylation. Taken together, we find that impaired cognition observed in NVH-lesioned rats is associated with decreased CaMKII and PKCα activities in memory-related brain regions, changes not rescued by risperidone treatment.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Transtornos Cognitivos/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Potenciação de Longa Duração/fisiologia , Inibição Neural/fisiologia , Proteína Quinase C-alfa/metabolismo , Animais , Animais Recém-Nascidos , Antipsicóticos/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/enzimologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Hipocampo/enzimologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Inibição Neural/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/enzimologia , Ratos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Risperidona/farmacologia
4.
Neuroscience ; 166(4): 1158-66, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20074623

RESUMO

Platelet-activating factor (PAF) is an important inflammatory lipid mediator affecting neural plasticity. In the present study, we demonstrated how PAF affects synaptic efficacy through activation of protein kinases in the rat hippocampal CA1 region. In cultured hippocampal neurons, 10 to 1000 nM PAF stimulated autophosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) and phosphorylation of synapsin I and myristoylated alanine-rich protein kinase C substrate (MARCKS). In hippocampal CA1 slices, field excitatory postsynaptic potentials (fEPSPs) induced by stimulation of the Schaffer collateral/commissural pathways were significantly increased 10-50 min after exposure to 100 to 1000 nM PAF. Immunoblotting analysis showed that 100 nM PAF treatment for 10 or 50 min significantly and persistently increased CaMKII autophosphorylation in the hippocampal CA1 region. Increased protein kinase Calpha (PKCalpha) autophosphorylation was also seen at the same time point after PAF exposure. By contrast, extracellular signal-regulated kinase (ERK) phosphorylation was slightly but significantly increased at 10 min after PAF exposure. Consistent with increased CaMKII autophosphorylation, AMPA-type glutamate receptor subunit 1 (GluR1) (Ser-831) phosphorylation as a CaMKII postsynaptic substrate significantly increased after 10 or 50 min of treatment, whereas synapsin I (Ser-603) phosphorylation as a presynaptic substrate increased at 10 min in the hippocampal CA1 region. Phosphorylation of MARCKS (Ser-152/156) and NMDA receptor subunit 1 (NR1) (Ser-896) as PKCalpha substrates also significantly increased after 10 min but had not further increased by 50 min in the CA1 region. Increased of fEPSPs induced by PAF treatment completely and/or partly inhibited by KN93 and/or U0126 treatment. These results suggest that PAF induces synaptic facilitation through activation of CaMKII, PKC and ERK in the hippocampal CA1 region.


Assuntos
Região CA1 Hipocampal/enzimologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Proteína Quinase C-alfa/metabolismo , Transmissão Sináptica/fisiologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Proteína Quinase C-alfa/efeitos dos fármacos , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Ratos , Ratos Wistar , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsinas/efeitos dos fármacos , Sinapsinas/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Fatores de Tempo
5.
Neuroscience ; 155(3): 876-87, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18616990

RESUMO

Although neurogenesis in the hippocampus is critical for improvement of depressive behaviors and cognitive functions in neurodegeneration disorders, there is no therapeutic agent available to promote neurogenesis in adult brain following brain ischemic injury. Here we found that i.p. administration of bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) [VO(OPT)], which stimulates phosphatidylinositol 3-kinase (PI3K)/Akt and extracellular signal regulated kinase (ERK) pathways, markedly enhanced brain ischemia-induced neurogenesis in the subgranular zone (SGZ) of the mouse hippocampus. VO(OPT) treatment enhanced not only the number of proliferating cells but also migration of neuroblasts. VO(OPT)-induced neurogenesis was associated with Akt and ERK activation in neural precursors in the SGZ. Likewise, VO(OPT)-induced neurogenesis was blocked by both PI3K/Akt and mitogen-activated protein kinase/extracellular signal regulated kinase kinase (MEK)/ERK inhibitors. VO(OPT) treatment rescued decreased phosphorylation of glycogen synthesis kinase 3beta (GSK-3beta) at Ser-9. Finally, amelioration of cognitive dysfunction seen following brain ischemia was positively correlated with VO(OPT)-induced neurogenesis. Taken together, VO(OPT) is a potential therapeutic agent that enhances ischemia-induced neurogenesis through PI3K/Akt and ERK activation, thereby improving memory and cognitive deficits following brain ischemia.


Assuntos
Isquemia Encefálica , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Compostos Organometálicos/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Bromodesoxiuridina/metabolismo , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
6.
Neuroscience ; 153(2): 414-27, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18400410

RESUMO

To define mechanisms underlying neurovascular injury following brain embolism-induced neurodegeneration, we investigated temporal and spatial pathological changes in brain microvessels up to 12 weeks after microsphere embolism (ME) induction in aged male rats. Mild ME upregulated endothelial nitric oxide synthase (eNOS) and protein tyrosine nitration in brain microvessels. Strong beta-amyloid immunoreactivity coincident with increased eNOS immunoreactivity was observed in microvessels. Immunoblotting of purified brain microvessels revealed that beta-amyloid accumulation significantly increased 1 week after ME induction and remained elevated for 12 weeks. Importantly, beta-amyloid accumulation in brain parenchyma was also observed in areas surrounding injured microvessels at 12 weeks. Levels of Alzheimer's-related hyperphosphorylated tau proteins also concomitantly increased in neurons surrounding regions of beta-amyloid accumulation 12 weeks after ME induction, as did glycogen synthase kinase (GSK3beta) (Tyr-216) phosphorylation. Taken together, ME-induced aberrant eNOS expression and subsequent protein tyrosine nitration in microvessels preceded beta-amyloid accumulation both in microvessels and brain parenchyma, leading to hyperphosphorylation of neuronal tau proteins through GSK3beta activation.


Assuntos
Envelhecimento/fisiologia , Peptídeos beta-Amiloides/metabolismo , Química Encefálica/fisiologia , Embolia/metabolismo , Microesferas , Proteínas tau/metabolismo , Animais , Western Blotting , Capilares/metabolismo , Circulação Cerebrovascular/fisiologia , Eletroforese em Gel de Poliacrilamida , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Imuno-Histoquímica , Masculino , Degeneração Neural/genética , Degeneração Neural/patologia , Óxido Nítrico Sintase Tipo III/biossíntese , Óxido Nítrico Sintase Tipo III/genética , Ácido Peroxinitroso/metabolismo , Fosforilação , Poli Adenosina Difosfato Ribose/metabolismo , Ratos , Ratos Wistar
7.
Neuroscience ; 151(3): 671-9, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18164824

RESUMO

The bilateral olfactory bulbectomy (OBX) mouse exhibits neurodegeneration of cholinergic neurons in the medial septum with concomitant cognitive deficits. Consistent with our previous observations, choline acetyltransferase (ChAT) protein levels in the medial septum decreased by 43.5% 2 weeks after OBX without changes in glutamic acid decarboxylase-65 (GAD65) levels. Interestingly, levels of the vesicular acetylcholine transporter (VAChT), which is localized at cholinergic neuron terminals, decreased both in hippocampal CA1 and CA3 regions following OBX. Confocal microscopy showed that VAChT expression was more severely reduced in CA3 14 days after OBX compared with CA1. Intriguingly, chronic treatment with a vanadium (IV) compound, VO(OPT) [bis(1-N-oxide-pyridine-2-thiolato)oxovanadium(IV)] (0.5-1 mg as vanadium (V)/kg/day, i.p.), significantly rescued cholinergic neurons in the medial septum in a dose-dependent manner. VO(OPT) treatment also prevented decreased VAChT immunoreactivity both in CA1 and CA3 regions in the hippocampus. Consistent with these findings, an impaired hippocampal long-term potentiation (LTP) and memory deficits seen in OBX mice were significantly prevented by VO(OPT) treatment. Taken together, OBX induces neurodegeneration of septo-hippocampal cholinergic neurons and impairment of memory-related behaviors. The neuroprotective effect of VO(OPT) could lead to novel therapeutic strategies to ameliorate cognitive deficits associated with cholinergic neuron degeneration in Alzheimer's disease and other neurodegenerative disorders.


Assuntos
Acetilcolina/metabolismo , Hipocampo/patologia , Degeneração Neural , Neurônios/efeitos dos fármacos , Bulbo Olfatório/lesões , Compostos Organometálicos/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Colina O-Acetiltransferase/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/patologia , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Técnicas de Patch-Clamp , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
8.
Neuroscience ; 148(1): 221-9, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17629407

RESUMO

We previously reported that orthovanadate composed of vanadate (V(5+)) activates phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling through inhibition of protein tyrosine phosphatases, thereby eliciting neuroprotection in brain ischemia/reperfusion injury. However, therapeutic doses of orthovanadate are associated with diarrhea due to inhibition of ATPase. By contrast, vanadyl (V(4+)) organic compounds show low cytotoxicity. Since both vanadate and vanadyl inhibit protein tyrosine phosphatases, we tested whether bis(1-oxy-2-pyridinethiolato)oxovanadium(IV) [VO(OPT)] in a vanadyl form elicits a neuroprotection in brain ischemia. In a mouse transient middle cerebral artery occlusion (MCAO) model, pre- and post-treatments with VO(OPT) significantly reduced infarct volume in a dose-dependent manner. Like orthovanadate, activation of the PI3K/Akt pathway mediated neuroprotective action. VO(OPT) treatment inhibited reduced Akt phosphorylation at Ser-473 following brain ischemia and restored decreased phosphorylation of forkhead box class O (FOXO) family members such as FKHR, FKHRL1, and AFX. Consistent with inhibition of FOXO dephosphorylation, VO(OPT) treatment blocked elevated expression of Fas-ligand, Bim and active caspase-3 24 h after ischemia/reperfusion. Taken together, a vanadyl compound, VO(OPT) elicits neuroprotective effects on brain ischemia/reperfusion injury without apparent side effects.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Vanadatos/farmacologia , Animais , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/enzimologia , Infarto Encefálico/fisiopatologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/fisiopatologia , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Proteína Ligante Fas/efeitos dos fármacos , Proteína Ligante Fas/metabolismo , Fatores de Transcrição Forkhead/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Masculino , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vanadatos/uso terapêutico
9.
Clin Pharmacol Ther ; 66(6): 569-81, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10613612

RESUMO

OBJECTIVE: To clarify the mechanism(s) for the interaction between warfarin and benzbromarone, a uricosuric agent, and to predict changes in the in vivo pharmacokinetics of (S)-warfarin from in vitro data. METHODS: Warfarin enantiomers and benzbromarone in serum, 7-hydroxywarfarin in urine, and serum unbound fractions of warfarin enantiomers were measured in patients with heart disease given warfarin with (n = 13) or without (n = 18) oral benzbromarone (50 mg/d). In vitro inhibition constants (K(i)) of benzbromarone for (S)-warfarin 7-hydroxylation were determined with use of human CYP2C9 and liver microsomes. The magnitude of changes in the formation clearance for 7-hydroxylation (CLf), the unbound oral clearance (CL(oral,u)), and the oral clearance (CL(oral)) for (S)-warfarin were predicted by equations incorporating the in vitro Ki, the theoretical maximum unbound hepatic benzbromarone concentration, and the fractions of warfarin eliminated through metabolism and of CYP2C9-mediated metabolic reaction susceptible to inhibition by benzbromarone. RESULTS: The patients given warfarin with benzbromarone required a 36% less (P < .01) warfarin dose than those given warfarin alone (2.5 versus 3.9 mg/d) to attain similar international normalized ratios (2.1 and 2.2, respectively), and the former had 65%, 53%, and 54% lower (P < .05 or P < .01) CLf, CL(oral),u, and CL(oral) for (S)-warfarin than the latter, respectively. In contrast, no significant differences were observed for (R)-warfarin kinetics between the groups. Benzbromarone was found to be a potent competitive inhibitor (Ki < 0.01 micromol/L) for (S)-warfarin 7-hydroxylation mediated by CYP2C9. The average changes in the in vivo CLf, CL(oral),u, and CL(oral)values for (S)-warfarin induced by benzbromarone were largely predictable by the proposed equations. CONCLUSION: Benzbromarone would intensify anticoagulant response of warfarin through an enantioselective inhibition of CYP2C9-mediated metabolism of pharmacologically more potent (S)-warfarin. The magnitude of changes in the in vivo warfarin kinetics may be predicted by in vitro data.


Assuntos
Anticoagulantes/farmacocinética , Hidrocarboneto de Aril Hidroxilases , Benzobromarona/farmacocinética , Esteroide 16-alfa-Hidroxilase , Uricosúricos/farmacocinética , Varfarina/farmacocinética , Idoso , Anticoagulantes/sangue , Anticoagulantes/urina , Benzobromarona/sangue , Benzobromarona/urina , Citocromo P-450 CYP2C9 , Sistema Enzimático do Citocromo P-450/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Masculino , Microssomos Hepáticos/enzimologia , Pessoa de Meia-Idade , Estereoisomerismo , Esteroide Hidroxilases/metabolismo , Uricosúricos/sangue , Uricosúricos/urina , Varfarina/sangue , Varfarina/urina
11.
FEBS Lett ; 355(1): 19-22, 1994 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-7957954

RESUMO

A variant of transforming growth factor-beta type II receptor (TGF-beta RII) cDNA was isolated from a mouse brain cDNA library. The predicted receptor is identical to previously reported mouse TGF-beta RII except that the isoform has an insertion sequence of 25 amino acids in the predicted ligand-binding domain. By the use of reverse transcription-polymerase chain reaction (RT-PCR), transcripts for both isoforms were detected in all tissues and developing embryos examined. The isoform transiently expressed in COS cells showed a similar ligand-binding specificity to authentic TGF-beta RII. These results suggest that the mouse TGF-beta RII gene generates multiple isoforms, possibly by alternative splicing, as reported for activin type IIB receptor; and an isoform which has the extra sequence in the ligand-binding domain is also involved in the TGF-beta signal transduction.


Assuntos
Clonagem Molecular , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Ligantes , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo
12.
Biochem Biophys Res Commun ; 198(3): 1063-9, 1994 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8117262
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...