Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Spec Care Dentist ; 40(5): 437-442, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32777084

RESUMO

AIM: To assess the efficacy of dexmedetomidine (DEX) on the intravenous moderate sedation (IVMS) regimen, while treating patients of the special patient care (SPC) population. This study aims to incorporate DEX into the typical IVMS drug regimen in order to reduce the amount of benzodiazepines (BZD) and opioids administered and as a result reduce the amount of unwanted side effects. METHOD AND RESULTS: A retrospective study was performed in the University of California Los Angeles (UCLA) SPC Clinic, where 42 patients were seen with and without DEX for dental treatment under IVMS. Medications administered, vital signs, and complications were recorded at 5 minute intervals over the first hour. All BZDs and opioids were converted to their IV midazolam and IV fentanyl equivalents, respectively. An opioid conversion equation was developed to summate the total amount of anesthetic agents administered. Data were analyzed by t-test. The amount of BZDs administered was reduced, however the decrease was not statistically significant (P = .066). There was a significant reduction in opioids (P < .05) and total anesthetic agents (P < .05) administered. CONCLUSION: The addition of DEX to the anesthetic regimen results in a reduction of overall medications administered.


Assuntos
Analgésicos Opioides , Dexmedetomidina , Benzodiazepinas , Sedação Consciente , Humanos , Hipnóticos e Sedativos , Los Angeles , Estudos Retrospectivos
2.
Toxicol Sci ; 122(1): 146-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21505090

RESUMO

Thalidomide (TD) displays remarkable species specificity, causing birth defects (teratogenesis) in humans and rabbits, but not rats or mice; yet, few determinants of species susceptibility have been identified. Also, certain mouse strains are susceptible to the embryopathic effects of some teratogens in embryo culture despite their resistance in vivo. Herein we show that CD-1 mouse embryos in culture are resistant to limb embryopathies caused by TD and two of its hydrolysis products, 2-phthalimidoglutaramic acid and 2-phthalimidoglutaric acid, although all three compounds cause these embryopathies in rabbit embryo culture. These results show that the resistance of CD-1 mice to TD teratogenesis is inherent to the embryo and is not dependent upon maternal factors, including differential in vivo exposure to the many hydrolysis products of TD. In utero TD exposure of rabbit but not mouse embryos elevates levels of the teratogenic oxidative DNA lesion 8-oxoguanine, which is repaired by oxoguanine glycosylase 1 (OGG1). However, DNA repair-deficient ogg1 knockout mice proved resistant to TD-initiated embryopathies in culture and teratogenesis in vivo, indicating that the resistance of mice is not due to a higher level of DNA repair.


Assuntos
DNA Glicosilases/genética , Distúrbios no Reparo do DNA/genética , Reparo do DNA/efeitos dos fármacos , Doenças Fetais/induzido quimicamente , Teratogênicos/toxicidade , Talidomida/toxicidade , Anormalidades Induzidas por Medicamentos , Animais , Dano ao DNA/efeitos dos fármacos , DNA Glicosilases/metabolismo , Distúrbios no Reparo do DNA/patologia , Técnicas de Cultura Embrionária , Feminino , Doenças Fetais/genética , Genótipo , Hidrólise , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Proteínas/efeitos dos fármacos
3.
Toxicol Sci ; 122(1): 157-69, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21505091

RESUMO

The expanding therapeutic uses of thalidomide (TD) are limited by its teratogenic side effects. Although the therapeutic and teratogenic effects may be stereoselectively separable, rapid in vivo racemization of the TD isomers confounds the corroboration of this distinction. Herein we evaluated the potential of fluorothalidomide (FTD), the closest structural analog of TD with stable, nonracemizing isomers, as a model compound for studying stereoselectivity in TD teratogenesis. In contrast to TD, FTD was a potent maternal and fetal toxicant in both rabbits and mice in vivo. Furthermore, FTD rapidly degraded in vivo, presumably via hydrolysis, which in vitro was up to 22-fold faster for FTD than TD. Most critically, in an established rabbit embryo culture model for TD teratogenesis, FTD did not initiate the limb bud embryopathies observed with TD. The chemical instability and strikingly different maternal and developmental toxicological profiles of FTD and TD make FTD an unsuitable compound for studying stereoselective mechanisms of TD teratogenesis.


Assuntos
Anormalidades Induzidas por Medicamentos/metabolismo , Embrião de Mamíferos/efeitos dos fármacos , Teratogênicos/toxicidade , Talidomida/toxicidade , Animais , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Técnicas de Cultura Embrionária , Feminino , Doenças Fetais/induzido quimicamente , Doenças Fetais/genética , Doenças Fetais/patologia , Hidrólise , Botões de Extremidades/anormalidades , Botões de Extremidades/efeitos dos fármacos , Masculino , Camundongos , Projetos Piloto , Gravidez , Coelhos
4.
FASEB J ; 25(7): 2468-83, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21502285

RESUMO

Thalidomide (TD) causes birth defects in humans and rabbits via several potential mechanisms, including bioactivation by embryonic prostaglandin H synthase (PHS) enzymes to a reactive intermediate that enhances reactive oxygen species (ROS) formation. We show herein that TD in rabbit embryo culture produces relevant embryopathies, including decreases in head/brain development by 28% and limb bud growth by 71% (P<0.05). Two TD hydrolysis products, 2-phthalimidoglutaramic acid (PGMA) and 2-phthalimidoglutaric acid (PGA), were similarly embryopathic, attenuating otic vesicle (ear) and limb bud formation by up to 36 and 77%, respectively (P<0.05). TD, PGMA, and PGA all increased embryonic DNA oxidation measured as 8-oxoguanine (8-oxoG) by up to 2-fold (P<0.05). Co- or pretreatment with the PHS inhibitors eicosatetraynoic acid (ETYA) or acetylsalicylic acid (ASA), or the free-radical spin trap phenylbutylnitrone (PBN), completely blocked embryonic 8-oxoG formation and/or embryopathies initiated by TD, PGMA, and PGA. This is the first demonstration of limb bud embryopathies initiated by TD, as well as its hydrolysis products, in a mammalian embryo culture model of a species susceptible to TD in vivo, indicating that all likely contribute to TD teratogenicity in vivo, in part through PHS-dependent, ROS-mediated mechanisms.


Assuntos
Embrião de Mamíferos/efeitos dos fármacos , Teratogênicos/toxicidade , Talidomida/toxicidade , Ácido 5,8,11,14-Eicosatetrainoico/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Animais , Aspirina/farmacologia , Encéfalo/anormalidades , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Feminino , Hidrólise , Botões de Extremidades/anormalidades , Botões de Extremidades/efeitos dos fármacos , Botões de Extremidades/metabolismo , Masculino , Estrutura Molecular , Oxirredução/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/metabolismo , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Teratogênicos/química , Teratogênicos/metabolismo , Talidomida/química , Talidomida/metabolismo , Técnicas de Cultura de Tecidos
5.
Handb Exp Pharmacol ; (196): 131-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20020262

RESUMO

Drugs and environmental chemicals can adversely alter the development of the fetus at critical periods during pregnancy, resulting in death, or in structural and functional birth defects in the surviving offspring. This process of teratogenesis may not be evident until a decade or more after birth. Postnatal functional abnormalities include deficits in brain function, a variety of metabolic diseases, and cancer. Due to the high degree of fetal cellular division and differentiation, and to differences from the adult in many biochemical pathways, the fetus is highly susceptible to teratogens, typically at low exposure levels that do not harm the mother. Insights into the mechanisms of teratogenesis come primarily from animal models and in vitro systems, and involve either receptor-mediated or reactive intermediate-mediated processes. Receptor-mediated mechanisms involving the reversible binding of xenobiotic substrates to a specific receptor are exemplified herein by the interaction of the environmental chemical 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD or "dioxin") with the cytosolic aryl hydrocarbon receptor (AHR), which translocates to the nucleus and, in association with other proteins, binds to AH-responsive elements (AHREs) in numerous genes, initiating changes in gene transcription that can perturb development. Alternatively, many xenobiotics are bioactivated by fetal enzymes like the cytochromes P450 (CYPs) and prostaglandin H synthases (PHSs) to highly unstable electrophilic or free radical reactive intermediates. Electrophilic reactive intermediates can covalently (irreversibly) bind to and alter the function of essential cellular macromolecules (proteins, DNA), causing developmental anomalies. Free radical reactive intermediates can enhance the formation of reactive oxygen species (ROS), resulting in oxidative damage to cellular macromolecules and/or altered signal transduction. The teratogenicity of reactive intermediates is determined to a large extent by the balance among embryonic and fetal pathways of xenobiotic bioactivation, detoxification of the xenobiotic reactive intermediate, detoxification of ROS, and repair of oxidative macromolecular damage.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Poluentes Ambientais/efeitos adversos , Morte Fetal/induzido quimicamente , Radicais Livres/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Teratogênicos/toxicidade , Anormalidades Induzidas por Medicamentos/metabolismo , Fatores Etários , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Poluentes Ambientais/metabolismo , Feminino , Morte Fetal/metabolismo , Feto/efeitos dos fármacos , Feto/metabolismo , Idade Gestacional , Humanos , Ligantes , Gravidez , Receptores de Hidrocarboneto Arílico/metabolismo , Medição de Risco
6.
Toxicol Sci ; 108(1): 4-18, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19126598

RESUMO

In the developing embryo and fetus, endogenous or xenobiotic-enhanced formation of reactive oxygen species (ROS) like hydroxyl radicals may adversely alter development by oxidatively damaging cellular lipids, proteins and DNA, and/or by altering signal transduction. The postnatal consequences may include an array of birth defects (teratogenesis), postnatal functional deficits, and diseases. In animal models, the adverse developmental consequences of in utero exposure to agents like thalidomide, methamphetamine, phenytoin, benzo[a]pyrene, and ionizing radiation can be modulated by altering pathways that control the embryonic ROS balance, including enzymes that bioactivate endogenous substrates and xenobiotics to free radical intermediates, antioxidative enzymes that detoxify ROS, and enzymes that repair oxidative DNA damage. ROS-mediated signaling via Ras, nuclear factor kappa B and related transducers also may contribute to altered development. Embryopathies can be reduced by free radical spin trapping agents and antioxidants, and enhanced by glutathione depletion. Further modulatory approaches to evaluate such mechanisms in vivo and/or in embryo culture have included the use of knockout mice, transgenic knock-ins and mutant deficient mice with altered enzyme activities, as well as antisense oligonucleotides, protein therapy with antioxidative enzymes, dietary depletion of essential cofactors and chemical enzyme inhibitors. In a few cases, measures anticipated to be protective have conversely enhanced the risk of adverse developmental outcomes, indicating the complexity of development and need for caution in testing therapeutic strategies in humans. A better understanding of the developmental effects of ROS may provide insights for risk assessment and the reduction of adverse postnatal consequences.


Assuntos
Malformações do Sistema Nervoso/induzido quimicamente , Estresse Oxidativo/fisiologia , Teratogênicos , Anormalidades Induzidas por Medicamentos , Animais , Dano ao DNA , Humanos , Modelos Animais , Neoplasias , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Xenobióticos
7.
Mol Cell Biol ; 27(4): 1425-32, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17158931

RESUMO

The stability and activity of tumor suppressor p53 are tightly regulated and partially depend on the p53 proline-rich domain (PRD). We recently analyzed mice expressing p53 with a deletion of the PRD (p53(DeltaP)). p53(DeltaP), a weak transactivator hypersensitive to Mdm2-mediated degradation, is unable to suppress oncogene-induced tumors. This phenotype could result from the loss of two motifs: Pin1 sites proposed to influence p53 stabilization and PXXP motifs proposed to mediate protein interactions. We investigated the importance of these motifs by generating mice encoding point mutations in the PRD. p53(TTAA) contains mutations suppressing all putative Pin1 sites in the PRD, while p53(AXXA) lacks PXXP motifs but retains one intact Pin1 site. Both mutant proteins accumulated in response to DNA damage, although the accumulation of p53(TTAA) was partially impaired. Importantly, p53(TTAA) and p53(AXXA) are efficient transactivators and potent suppressors of oncogene-induced tumors. Thus, Pin1 sites in the PRD may modulate p53 stability but do not significantly affect function. In addition, PXXP motifs are not essential, but structure dictated by the presence of prolines, PXXXXP motifs that may mediate protein interactions, and/or the length of this region appears to be functionally significant. These results may explain why the sequence of the p53 PRD is so variable in evolution.


Assuntos
Neoplasias/patologia , Prolina/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Sítios de Ligação , Ciclo Celular , Proliferação de Células , Transformação Celular Neoplásica , Sequência Conservada , Dano ao DNA , Fibroblastos/citologia , Marcação de Genes , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Mutação Puntual/genética , Ligação Proteica , Estrutura Terciária de Proteína , Recombinação Genética/genética , Relação Estrutura-Atividade , Ativação Transcricional/genética
8.
Nucleic Acids Res ; 34(13): e92, 2006 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16870721

RESUMO

In recent years, tremendous insight has been gained on p53 regulation by targeting mutations at the p53 locus using homologous recombination in ES cells to generate mutant mice. Although informative, this approach is inefficient, slow and expensive. To facilitate targeting at the p53 locus, we developed an improved Recombinase-Mediated Cassette Exchange (RMCE) method. Our approach enables efficient targeting in ES cells to facilitate the production of mutant mice. But more importantly, the approach was Adapted for targeting in Somatic cells to Accelerate Phenotyping (RMCE-ASAP). We provide proof-of-concept for this at the p53 locus, by showing efficient targeting in fibroblasts, and rapid phenotypic read-out of a recessive mutation after a single exchange. RMCE-ASAP combines inverted heterologous recombinase target sites, a positive/negative selection marker that preserves the germline capacity of ES cells, and the power of mouse genetics. These general principles should make RMCE-ASAP applicable to any locus.


Assuntos
Embrião de Mamíferos/citologia , Marcação de Genes/métodos , Genes p53 , Camundongos/genética , Animais , Células Cultivadas , Integrases , Fenótipo , Recombinação Genética , Células-Tronco , Proteínas Virais
9.
Cancer Cell ; 9(4): 273-85, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16616333

RESUMO

The mechanisms by which Mdm2 and Mdm4 (MdmX) regulate p53 remain controversial. We generated a mouse encoding p53 lacking the proline-rich domain (p53DeltaP). p53DeltaP exhibited increased sensitivity to Mdm2-dependent degradation and decreased transactivation capacity, correlating with deficient cell cycle arrest and reduced apoptotic responses. p53DeltaP induced lethality in Mdm2-/- embryos, but not in Mdm4-/- embryos. Mdm4 loss did not alter Mdm2 stability but significantly increased p53DeltaP transactivation to partially restore cycle control. In contrast, decreasing Mdm2 levels increased p53DeltaP levels without altering p53DeltaP transactivation. Thus, Mdm4 regulates p53 activity, while Mdm2 mainly controls p53 stability. Furthermore, Mdm4 loss dramatically improved p53DeltaP-mediated suppression of oncogene-induced tumors, emphasizing the importance of targeting Mdm4 in chemotherapies designed to activate p53.


Assuntos
Mutação/genética , Prolina/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Células Cultivadas , DNA/genética , Dano ao DNA , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Prolina/genética , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2/deficiência , Proteínas Proto-Oncogênicas c-mdm2/genética , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/química , Ubiquitina-Proteína Ligases/genética
10.
Proc Natl Acad Sci U S A ; 102(29): 10188-93, 2005 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-16006521

RESUMO

P53 is an unstable transcription factor that is mutated in a majority of human cancers. With a significant role in initiating cell elimination programs, a network has evolved to fine-tune P53 transcriptional output and prevent errant activation. Modifications of the C terminus have long been viewed as critical binary determinants of P53 stability or activation. However, these conclusions are based on in vitro transfection or biochemical analyses where the stoichiometries between P53 and its regulators are perturbed. Therefore, we tested the importance of the C-terminal regulatory region for P53 control in mice where the seven C-terminal lysines were changed to arginine (Trp-53(7KR)). Surprisingly, the homozygous mutant mice are viable and phenotypically normal. We have functionally characterized the mutant protein in both MEFs and thymocytes, revealing the unexpected result that Trp-53(7KR) exhibits a normal half-life and functions like WT P53 in cell cycle arrest and apoptosis, and in an E1A-ras xenograft tumor suppression assay. However, a significant difference is that P53(7KR) is activated more easily by DNA damage in thymus than WT P53. Importantly, although MEFs encoding WT P53 spontaneously emerge from crisis to become immortal in a 3T3 growth protocol, we do not observe any such escape with the P53(7KR) cells. We propose that the C-terminal modifications believed to be critical for proper P53 regulation are not essential, but contribute to a fine-tuning mechanism of homeostatic control in vivo.


Assuntos
Dano ao DNA , Regulação da Expressão Gênica/genética , Lisina/genética , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Sequência de Aminoácidos , Animais , Apoptose/genética , Western Blotting , Ciclo Celular/genética , Primers do DNA , Citometria de Fluxo , Imunofluorescência , Genótipo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Mutação/genética , Timo/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...