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

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Cell ; 30(5): 543-4, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18538651

RESUMO

In a recent issue of Molecular Cell, Enquist-Newman et al. (2008) demonstrate that Acm1 is ubiquitinated by APC(Cdc20). By contrast, the high-affinity interaction between Acm1 and APC(Cdh1) renders it a poor substrate, but a specific inhibitor, of the APC(Cdh1) complex.


Assuntos
Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Proteínas Cdc20 , Proteínas Cdh1 , Proteínas de Ciclo Celular/metabolismo , Mitose , Especificidade por Substrato , Ubiquitinação
2.
Curr Pharm Teach Learn ; 16(3): 221-230, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38281827

RESUMO

BACKGROUND: Clinical pharmacogenomics is an expanding area in healthcare that relies heavily on pharmacists for advocacy and implementation. To support pharmacists' significant roles in clinical pharmacogenomics, pharmacy schools and colleges in the United States (US) have strived to incorporate pharmacogenomics education into their curricula, and various teaching strategies have been employed in recent years to meet pharmacogenomics educational outcomes. The six major strategies reported in the literature are described and compared in this review, which culminates in a proposed longitudinal curriculum design for pharmacogenomics education. METHODS: Publications focused on pharmacogenomics education to pharmacy students within the US in the past decade were evaluated and summarized. RESULTS: The major education strategies that have been studied are didactic lecture, personal genotyping or personal genomic testing, simulation laboratory activity, interprofessional education, practice-based activity such as clinical rotation, and combinational courses. Strengths and limitations of each teaching strategy are summarized and discussed. IMPLICATIONS: Based upon each education strategy's strengths and weaknesses, the authors propose a longitudinal curriculum design to ensure that pharmacogenomics is taught multiple times to pharmacy students with diverse formats and teaching objectives conducive to long-term knowledge retention and practice readiness. Through this longitudinal curriculum design, pharmacy graduates will be well equipped to lead clinical pharmacogenomics in practice.


Assuntos
Educação em Farmácia , Farmacogenética , Estados Unidos , Humanos , Farmacogenética/educação , Faculdades de Farmácia , Currículo , Farmacêuticos
3.
Curr Biol ; 18(13): 933-42, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18571408

RESUMO

BACKGROUND: In response to DNA damage, cells undergo either cell-cycle arrest or apoptosis, depending on the extent of damage and the cell's capacity for DNA repair. Cell-cycle arrest induced by double-stranded DNA breaks depends on activation of the ataxia-telangiectasia (ATM) protein kinase, which phosphorylates cell-cycle effectors such as Chk2 and p53 to inhibit cell-cycle progression. ATM is recruited to double-stranded DNA breaks by a complex of sensor proteins, including Mre11/Rad50/Nbs1, resulting in autophosphorylation, monomerization, and activation of ATM kinase. RESULTS: In characterizing Aven protein, a previously reported apoptotic inhibitor, we have found that Aven can function as an ATM activator to inhibit G2/M progression. Aven bound to ATM and Aven overexpressed in cycling Xenopus egg extracts prevented mitotic entry and induced phosphorylation of ATM and its substrates. Immunodepletion of endogenous Aven allowed mitotic entry even in the presence of damaged DNA, and RNAi-mediated knockdown of Aven in human cells prevented autophosphorylation of ATM at an activating site (S1981) in response to DNA damage. Interestingly, Aven is also a substrate of the ATM kinase. Mutation of ATM-mediated phosphorylation sites on Aven reduced its ability to activate ATM, suggesting that Aven activation of ATM after DNA damage is enhanced by ATM-mediated Aven phosphorylation. CONCLUSIONS: These results identify Aven as a new ATM activator and describe a positive feedback loop operating between Aven and ATM. In aggregate, these findings place Aven, a known apoptotic inhibitor, as a critical transducer of the DNA-damage signal.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Ativação Enzimática , Retroalimentação Fisiológica , Células HeLa , Humanos , Fosforilação , Interferência de RNA , Xenopus
4.
J Pharm Sci ; 108(12): 3870-3878, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31545969

RESUMO

Eflornithine has been used to treat second-stage human African trypanosomiasis. However, it has inadequate oral bioavailability and low blood-brain barrier permeation, thus requiring a lengthy and complicated intravenous infusion schedule. Here, we investigated the feasibility of using an intercellular junction-modulating E-cadherin peptide HAV6 to enhance the oral bioavailability and blood-brain barrier permeation of eflornithine. Eflornithine was not metabolized in liver microsomes, nor was it a substrate for the human efflux transporter P-glycoprotein. Furthermore, HAV6 and HAV6scr (sequence scrambled HAV6) were stable in simulated gastric fluid with pepsin and rat intestinal mucosal scrapings. Both peptides were stable in human plasma, albeit less stable in rat and mouse plasma. HAV6 increased eflornithine permeability across Madin-Darby canine kidney and Caco-2 cell monolayers (5- and up to 8.5-fold), whereas HAV6scr had little effect. Using an in situ rat brain perfusion model, HAV6, but not HAV6scr, significantly increased eflornithine concentrations in different brain regions up to 4.9-fold. In rats, coadministration of HAV6 increased eflornithine oral bioavailability from 38% to 54%, brain concentrations by up to 83%, and cerebrospinal fluid concentrations by 40%. In conclusion, coadministration of HAV6, either during intravenous infusion or as an oral formulation, has the potential to improve eflornithine-based treatment for second-stage human African trypanosomiasis.


Assuntos
Encéfalo/metabolismo , Caderinas/metabolismo , Eflornitina/metabolismo , Junções Intercelulares/metabolismo , Peptídeos/metabolismo , Administração Oral , Animais , Disponibilidade Biológica , Transporte Biológico/fisiologia , Barreira Hematoencefálica/metabolismo , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Cães , Humanos , Células Madin Darby de Rim Canino , Masculino , Permeabilidade , Ratos , Ratos Sprague-Dawley
5.
Proteomics ; 8(20): 4186-96, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18792928

RESUMO

The human cytochrome P450 (P450) superfamily consists of membrane-bound proteins that metabolize a myriad of xenobiotics and endogenous compounds. Quantification of P450 expression in various tissues under normal and induced conditions has an important role in drug safety and efficacy. Conventional immunoquantification methods have poor dynamic range, low throughput, and a limited number of specific antibodies. Recent advances in MS-based quantitative proteomics enable absolute protein quantification in a complex biological mixture. We have developed a gel-free MS-based protein quantification strategy to quantify CYP3A enzymes in human liver microsomes (HLM). Recombinant protein-derived proteotypic peptides and synthetic stable isotope-labeled proteotypic peptides were used as calibration standards and internal standards, respectively. The lower limit of quantification was approximately 20 fmol P450. In two separate panels of HLM examined (n = 11 and n = 22), CYP3A, CYP3A4 and CYP3A5 concentrations were determined reproducibly (CV or=0.87) and marker activities (r(2)>or=0.88), including testosterone 6beta-hydroxylation (CYP3A), midazolam 1'-hydroxylation (CYP3A), itraconazole 6-hydroxylation (CYP3A4) and CYP3A5-mediated vincristine M1 formation (CYP3A5). Taken together, our MS-based method provides a specific, sensitive and reliable means of P450 protein quantification and should facilitate P450 characterization during drug development, especially when specific substrates and/or antibodies are unavailable.


Assuntos
Sistema Enzimático do Citocromo P-450/análise , Espectrometria de Massas/métodos , Microssomos Hepáticos/química , Proteômica/métodos , Adolescente , Adulto , Idoso , Criança , Citocromo P-450 CYP3A , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Homologia de Sequência de Aminoácidos , Incerteza
6.
J Biomol Screen ; 21(10): 1090-1099, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28139960

RESUMO

Cytochrome P450 1B1 (CYP1B1) is an anticancer therapeutic target due to its overexpression in a number of steroid hormone-related cancers. One anticancer drug discovery strategy is to develop prodrugs specifically activated by CYP1B1 in malignant tissues to cytotoxic metabolites. Here, we aimed to develop an in vitro screening model for CYP1B1-targeted anticancer prodrugs using the KLE human endometrial carcinoma cell line. KLE cells demonstrated superior stability of CYP1B1 expression relative to transiently transfected cells and did not express any appreciable amount of cognate CYP1A1 or CYP1A2, which would have compromised the specificity of the screening assay. The effect of two CYP1B1-targeted probe prodrugs on KLE cells was evaluated in the absence and presence of a CYP1B1 inhibitor to chemically "knock out" CYP1B1 activity (CYP1B1 inhibited). Both probe prodrugs were more toxic to KLE cells than to CYP1B1-inhibited KLE cells and significantly induced G0/G1 arrest and decreased the S phase in KLE cells. They also exhibited pro-apoptotic effects in KLE cells, which were attenuated in CYP1B1-inhibited KLE cells. In summary, a KLE cell-based model has been characterized to be suitable for identifying CYP1B1-targeted anticancer prodrugs and should be further developed and employed for screening chemical libraries.


Assuntos
Antineoplásicos/isolamento & purificação , Citocromo P-450 CYP1B1/antagonistas & inibidores , Ensaios de Triagem em Larga Escala/métodos , Neoplasias/tratamento farmacológico , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocromo P-450 CYP1B1/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Marcação de Genes/métodos , Humanos , Terapia de Alvo Molecular , Pró-Fármacos/isolamento & purificação , Pró-Fármacos/farmacologia
7.
Mol Biol Cell ; 21(15): 2589-97, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20534816

RESUMO

Vertebrate eggs are arrested at Metaphase II by Emi2, the meiotic anaphase-promoting complex/cyclosome (APC/C) inhibitor. Although the importance of Emi2 during oocyte maturation has been widely recognized and its regulation extensively studied, its mechanism of action remained elusive. Many APC/C inhibitors have been reported to act as pseudosubstrates, inhibiting the APC/C by preventing substrate binding. Here we show that a previously identified zinc-binding region is critical for the function of Emi2, whereas the D-box is largely dispensable. We further demonstrate that instead of acting through a "pseudosubstrate" mechanism as previously hypothesized, Emi2 can inhibit Cdc20-dependent activation of the APC/C substoichiometrically, blocking ubiquitin transfer from the ubiquitin-charged E2 to the substrate. These findings provide a novel mechanism of APC/C inhibition wherein the final step of ubiquitin transfer is targeted and raise the interesting possibility that APC/C is inhibited by Emi2 in a catalytic manner.


Assuntos
Proteínas F-Box/química , Proteínas F-Box/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitina/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Motivos de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Animais , Biocatálise , Ativação Enzimática , Humanos , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores
8.
Nat Cell Biol ; 11(5): 644-51, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19396163

RESUMO

Loss of cell division cycle 2 (Cdc2, also known as Cdk1) activity after cyclin B degradation is necessary, but not sufficient, for mitotic exit. Proteins phosphorylated by Cdc2 and downstream mitotic kinases must be dephosphorylated. We report here that protein phosphatase-1 (PP1) is the main catalyst of mitotic phosphoprotein dephosphorylation. Suppression of PP1 during early mitosis is maintained through dual inhibition by Cdc2 phosphorylation and the binding of inhibitor-1. Protein kinase A (PKA) phosphorylates inhibitor-1, mediating binding to PP1. As Cdc2 levels drop after cyclin B degradation, auto-dephosphorylation of PP1 at its Cdc2 phosphorylation site (Thr 320) allows partial PP1 activation. This promotes PP1-regulated dephosphorylation at the activating site of inhibitor-1 (Thr 35) followed by dissociation of the inhibitor-1-PP1 complex and then full PP1 activation to promote mitotic exit. Thus, Cdc2 both phosphorylates multiple mitotic substrates and inhibits their PP1-mediated dephosphorylation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mitose/fisiologia , Fosfoproteínas/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Proteína Quinase CDC2 , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclina B/metabolismo , Ciclina B/farmacologia , Quinases Ciclina-Dependentes , Células HeLa , Humanos , Modelos Biológicos , Ácido Okadáico/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação , Ligação Proteica/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/farmacologia , Proteínas/farmacologia , Purinas/farmacologia , Roscovitina , Treonina/metabolismo , Proteínas de Xenopus/antagonistas & inibidores , Proteínas de Xenopus/metabolismo , Xenopus laevis
9.
J Cell Sci ; 121(Pt 21): 3509-14, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18946022

RESUMO

Vertebrate eggs are arrested at the metaphase stage of meiosis II. Only upon fertilization will the metaphase-II-arrested eggs exit meiosis II and enter interphase. In 1971, Masui and Markert injected egg extracts into a two-cell-stage embryo and found that the injected blastomere arrested at the next mitosis. On the basis of these observations, they proposed the existence of an activity present in the eggs that is responsible for meiosis-II arrest and can induce mitotic arrest, and named this activity cytostatic factor (CSF). Although the existence of CSF was hypothesized more than 35 years ago, its precise identity remained unclear until recently. The discovery of the Mos-MAPK pathway and characterization of the anaphase-promoting complex/cyclosome (APC/C) as a central regulator of M-phase exit provided the framework for a molecular understanding of CSF. These pathways have now been linked by the discovery and characterization of the protein Emi2, a meiotic APC/C inhibitor, the activity and stability of which are controlled by the Mos-MAPK pathway. Continued investigation into the mechanism of action and mode of regulation of Emi2 promises to shed light not only on CSF function, but also on the general principles of APC/C regulation and the control of protein function by MAPK pathways.


Assuntos
Ciclina B/metabolismo , Proteínas F-Box/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Meiose , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Biologia do Desenvolvimento , Retroalimentação Fisiológica , Sistema de Sinalização das MAP Quinases , Mitose , Modelos Biológicos , Oócitos/metabolismo , Fosforilação
10.
Mol Biol Cell ; 19(8): 3536-43, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550795

RESUMO

The transition of oocytes from meiosis I (MI) to meiosis II (MII) requires partial cyclin B degradation to allow MI exit without S phase entry. Rapid reaccumulation of cyclin B allows direct progression into MII, producing a cytostatic factor (CSF)-arrested egg. It has been reported that dampened translation of the anaphase-promoting complex (APC) inhibitor Emi2 at MI allows partial APC activation and MI exit. We have detected active Emi2 translation at MI and show that Emi2 levels in MI are mainly controlled by regulated degradation. Emi2 degradation in MI depends not on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), but on Cdc2-mediated phosphorylation of multiple sites within Emi2. As in MII, this phosphorylation is antagonized by Mos-mediated recruitment of PP2A to Emi2. Higher Cdc2 kinase activity in MI than MII allows sufficient Emi2 phosphorylation to destabilize Emi2 in MI. At MI anaphase, APC-mediated degradation of cyclin B decreases Cdc2 activity, enabling Cdc2-mediated Emi2 phosphorylation to be successfully antagonized by Mos-mediated PP2A recruitment. These data suggest a model of APC autoinhibition mediated by stabilization of Emi2; Emi2 proteins accumulate at MI exit and inhibit APC activity sufficiently to prevent complete degradation of cyclin B, allowing MI exit while preventing interphase before MII entry.


Assuntos
Ciclina B/fisiologia , Proteínas F-Box/fisiologia , Regulação da Expressão Gênica , Meiose , Proteínas Proto-Oncogênicas c-mos/fisiologia , Animais , Proteína Quinase CDC2 , Movimento Celular , Ciclina B/metabolismo , Quinases Ciclina-Dependentes , Endocitose , Células HL-60 , Humanos , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo
11.
Drug Metab Dispos ; 35(11): 2067-75, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17709372

RESUMO

CYP4F enzymes, including CYP4F2 and CYP4F3B, were recently shown to be the major enzymes catalyzing the initial oxidative O-demethylation of the antiparasitic prodrug pafuramidine (DB289) by human liver microsomes. As suggested by a low oral bioavailability, DB289 could undergo first-pass biotransformation in the intestine, as well as in the liver. Using human intestinal microsomes (HIM), we characterized the enteric enzymes that catalyze the initial O-demethylation of DB289 to the intermediate metabolite, M1. M1 formation in HIM was catalyzed by cytochrome P450 (P450) enzymes, as evidenced by potent inhibition by 1-aminobenzotriazole and the requirement for NADPH. Apparent K(m) and V(max) values ranged from 0.6 to 2.4 microM and from 0.02 to 0.89 nmol/min/mg protein, respectively (n = 9). Of the P450 chemical inhibitors evaluated, ketoconazole was the most potent, inhibiting M1 formation by 66%. Two inhibitors of P450-mediated arachidonic acid metabolism, HET0016 (N-hydroxy-N'-(4-n-butyl-2-methylphenyl)formamidine) and 17-octadecynoic acid, inhibited M1 formation in a concentration-dependent manner (up to 95%). Immunoinhibition with an antibody raised against CYP4F2 showed concentration-dependent inhibition of M1 formation (up to 92%), whereas antibodies against CYP3A4/5 and CYP2J2 had negligible to modest effects. M1 formation rates correlated strongly with arachidonic acid omega-hydroxylation rates (r(2) = 0.94, P < 0.0001, n = 12) in a panel of HIM that lacked detectable CYP4A11 protein expression. Quantitative Western blot analysis revealed appreciable CYP4F expression in these HIM, with a mean (range) of 7 (3-18) pmol/mg protein. We conclude that enteric CYP4F enzymes could play a role in the first-pass biotransformation of DB289 and other xenobiotics.


Assuntos
Benzamidinas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Intestinos/enzimologia , Microssomos/enzimologia , Pró-Fármacos/metabolismo , Amidinas/farmacologia , Anticorpos/farmacologia , Antiparasitários/química , Antiparasitários/metabolismo , Antiparasitários/farmacocinética , Ácido Araquidônico/metabolismo , Ácido Araquidônico/farmacologia , Benzamidinas/química , Benzamidinas/farmacocinética , Benzoflavonas/farmacologia , Butirofenonas/farmacologia , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP2J2 , Citocromo P-450 CYP3A , Citocromo P-450 CYP4A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/imunologia , Família 4 do Citocromo P450 , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Mucosa Intestinal/metabolismo , Cinética , Metilação/efeitos dos fármacos , Microssomos/metabolismo , Oxigenases/antagonistas & inibidores , Oxigenases/imunologia , Oxigenases/metabolismo , Piperidinas/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Proteínas Recombinantes/metabolismo , Estereoisomerismo
12.
Proc Natl Acad Sci U S A ; 104(42): 16564-9, 2007 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17881560

RESUMO

Before fertilization, vertebrate eggs are arrested in meiosis II by cytostatic factor (CSF), which holds the anaphase-promoting complex (APC) in an inactive state. It was recently reported that Mos, an integral component of CSF, acts in part by promoting the Rsk-mediated phosphorylation of the APC inhibitor Emi2/Erp1. We report here that Rsk phosphorylation of Emi2 promotes its interaction with the protein phosphatase PP2A. Emi2 residues adjacent to the Rsk phosphorylation site were important for PP2A binding. An Emi2 mutant that retained Rsk phosphorylation but lacked PP2A binding could not be modulated by Mos. PP2A bound to Emi2 acted on two distinct clusters of sites phosphorylated by Cdc2, one responsible for modulating its stability during CSF arrest and one that controls binding to the APC. These findings provide a molecular mechanism for Mos action in promoting CSF arrest and also define an unusual mechanism, whereby protein phosphorylation recruits a phosphatase for dephosphorylation of distinct sites phosphorylated by another kinase.


Assuntos
Proteínas F-Box/metabolismo , Meiose , Óvulo/fisiologia , Fosforilase Fosfatase/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas de Xenopus/metabolismo , Sequência de Aminoácidos , Animais , Proteínas F-Box/genética , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-mos/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Xenopus , Proteínas de Xenopus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA