Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Pharmacol Sin ; 43(10): 2527-2541, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35347247

RESUMO

Oxidative stress is extensively involved in neurodegeneration. Clinical evidence shows that keeping the mind active through mentally-stimulating physical activities can effectively slow down the progression of neurodegeneration. With increased physical activities, more neurotransmitters would be released in the brain. In the present study, we investigated whether some of the released neurotransmitters might have a beneficial effect against oxidative neurodegeneration in vitro. Glutamate-induced, glutathione depletion-associated oxidative cytotoxicity in HT22 mouse hippocampal neuronal cells was used as an experimental model. We showed that norepinephrine (NE, 50 µM) or dopamine (DA, 50 µM) exerted potent protective effect against glutamate-induced cytotoxicity, but this effect was not observed when other neurotransmitters such as histamine, γ-aminobutyric acid, serotonin, glycine and acetylcholine were tested. In glutamate-treated HT22 cells, both NE and DA significantly suppressed glutathione depletion-associated mitochondrial dysfunction including mitochondrial superoxide accumulation, ATP depletion and mitochondrial AIF release. Moreover, both NE and DA inhibited glutathione depletion-associated MAPKs activation, p53 phosphorylation and GADD45α activation. Molecular docking analysis revealed that NE and DA could bind to protein disulfide isomerase (PDI). In biochemical enzymatic assay in vitro, NE and DA dose-dependently inhibited the reductive activity of PDI. We further revealed that the protective effect of NE and DA against glutamate-induced oxidative cytotoxicity was mediated through inhibition of PDI-catalyzed dimerization of the neuronal nitric oxide synthase. Collectively, the results of this study suggest that NE and DA may have a protective effect against oxidative neurodegeneration through inhibition of protein disulfide isomerase and the subsequent activation of the MAPKs‒p53‒GADD45α oxidative cascade.


Assuntos
Morte Celular , Dopamina , Neuroproteção , Norepinefrina , Isomerases de Dissulfetos de Proteínas , Acetilcolina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Dopamina/farmacologia , Ácido Glutâmico/metabolismo , Glutationa/metabolismo , Glicina/farmacologia , Histamina/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Neuroproteção/efeitos dos fármacos , Neurotransmissores , Óxido Nítrico Sintase Tipo I/metabolismo , Norepinefrina/farmacologia , Estresse Oxidativo , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Serotonina/metabolismo , Serotonina/farmacologia , Superóxidos/metabolismo , Superóxidos/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Ácido gama-Aminobutírico/metabolismo
2.
Food Funct ; 11(8): 7255-7265, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32776051

RESUMO

d-Limonene, a type of natural extract obtained from citrus oils, was reported to have anti-cancer effects and be well-tolerated by cancer patients. Despite arousing interest as a cancer chemopreventive substance, the transcriptomic profile of d-limonene in humans is poorly understood. Based on the results of the transcriptomic profiling, a lncRNA named protein disulfide isomerase family A member three pseudogene (PDIA3P1) was found to be regulated by d-limonene. PDIA3P1 is an oncogene verified by three lung adenocarcinoma (LUAD) datasets. The knockdown of PDIA3P1 with siRNA decreased the viability, invasion, migration, and proliferation of LUAD cells. Based on The Cancer Genome Atlas (TCGA) LUAD datasets, PDIA3P1 regulates functions and pathways mainly including lipid metabolism, immunity, and the change of the chromosome structure. This study comprehensively performs the transcriptomic analysis of the d-limonene regulation on LUAD, and reveals that PDIA3P1 may be the mediator in helping d-limonene to prevent and suppress LUAD via lipid metabolism, immunity pathway, and the change in the chromosome structure.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Citrus/química , Limoneno/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Óleos de Plantas/farmacologia , Adenocarcinoma/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Pseudogenes/efeitos dos fármacos , RNA Longo não Codificante/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia
3.
Hepatology ; 66(3): 855-868, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439950

RESUMO

Sorafenib is the only approved targeted drug for hepatocellular carcinoma (HCC), but its effect on patients' survival gain is limited and varies over a wide range depending on pathogenetic conditions. Thus, enhancing the efficacy of sorafenib and finding a reliable predictive biomarker are crucial to achieve efficient control of HCCs. In this study, we utilized a systems approach by combining transcriptome analysis of the mRNA changes in HCC cell lines in response to sorafenib with network analysis to investigate the action and resistance mechanism of sorafenib. Gene list functional enrichment analysis and gene set enrichment analysis revealed that proteotoxic stress and apoptosis modules are activated in the presence of sorafenib. Further analysis of the endoplasmic reticulum stress network model, combined with in vitro experiments, showed that introducing an additional stress by treating the orally active protein disulfide isomerase (PDI) inhibitor (PACMA 31) can synergistically increase the efficacy of sorafenib in vitro and in vivo, which was confirmed using a mouse xenograft model. We also found that HCC patients with high PDI expression show resistance to sorafenib and poor clinical outcomes, compared to the low-PDI-expression group. CONCLUSION: These results suggest that PDI is a promising therapeutic target for enhancing the efficacy of sorafenib and can also be a biomarker for predicting sorafenib responsiveness. (Hepatology 2017;66:855-868).


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Niacinamida/administração & dosagem , Modelos de Riscos Proporcionais , Isomerases de Dissulfetos de Proteínas/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Distribuição Aleatória , Sorafenibe , Estatísticas não Paramétricas , Células Tumorais Cultivadas
4.
JCI Insight ; 2(1): e89373, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28097231

RESUMO

BACKGROUND: Protein disulfide isomerase (PDI) is required for thrombus formation. We previously demonstrated that glycosylated quercetin flavonoids such as isoquercetin inhibit PDI activity and thrombus formation in animal models, but whether extracellular PDI represents a viable anticoagulant target in humans and how its inhibition affects blood coagulation remain unknown. METHODS: We evaluated effects of oral administration of isoquercetin on platelet-dependent thrombin generation in healthy subjects and patients with persistently elevated anti-phospholipid antibodies. RESULTS: Following oral administration of 1,000 mg isoquercetin to healthy adults, the measured peak plasma quercetin concentration (9.2 µM) exceeded its IC50 for inhibition of PDI by isoquercetin in vitro (2.5 ± 0.4 µM). Platelet-dependent thrombin generation decreased by 51% in the healthy volunteers compared with baseline (P = 0.0004) and by 64% in the anti-phospholipid antibody cohort (P = 0.015) following isoquercetin ingestion. To understand how PDI affects thrombin generation, we evaluated substrates of PDI identified using an unbiased mechanistic-based substrate trapping approach. These studies identified platelet factor V as a PDI substrate. Isoquercetin blocked both platelet factor Va and thrombin generation with an IC50 of ~5 µM. Inhibition of PDI by isoquercetin ingestion resulted in a 53% decrease in the generation of platelet factor Va (P = 0.001). Isoquercetin-mediated inhibition was reversed with addition of exogenous factor Va. CONCLUSION: These studies show that oral administration of isoquercetin inhibits PDI activity in plasma and diminishes platelet-dependent thrombin generation predominantly by blocking the generation of platelet factor Va. These pharmacodynamic and mechanistic observations represent an important step in the development of a novel class of antithrombotic agents targeting PDI. TRIAL REGISTRATION: Clinicaltrials.gov (NCT01722669) FUNDING: National Heart, Lung, and Blood Institute (U54 HL112302) and Quercegen Pharma.


Assuntos
Fatores de Coagulação Sanguínea/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Quercetina/análogos & derivados , Trombina/efeitos dos fármacos , Administração Oral , Animais , Síndrome Antifosfolipídica/imunologia , Humanos , Modelos Animais , Quercetina/administração & dosagem , Quercetina/farmacologia
5.
Respir Res ; 14: 141, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24364984

RESUMO

BACKGROUND: The endoplasmic reticulum (ER) stress response participates in many chronic inflammatory and autoimmune diseases. In the current study, we sought to examine the contribution of ER stress transducers in the pathogenesis of three principal facets of allergic asthma: inflammation, airway fibrosis, and airways hyperresponsiveness. METHODS: House Dust Mite (HDM) was used as an allergen for in vitro and in vivo challenge of primary human and murine airway epithelial cells. ER stress transducers were modulated using specific small interfering RNAs (siRNAs) in vivo. Inflammation, airway remodeling, and hyperresponsiveness were measured by total bronchoalveolar lavage (BAL) cell counts, determination of collagen, and methacholine responsiveness in mice, respectively. RESULTS: Challenge of human bronchiolar and nasal epithelial cells with HDM extract induced the ER stress transducer, activating transcription factor 6 α (ATF6α) as well as protein disulfide isomerase, ERp57, in association with activation of caspase-3. SiRNA-mediated knockdown of ATF6α and ERp57 during HDM administration in mice resulted in a decrease in components of HDM-induced ER stress, disulfide mediated oligomerization of Bak, and activation of caspase-3. Furthermore, siRNA-mediated knockdown of ATF6α and ERp57 led to decreased inflammation, airway hyperresponsiveness and airway fibrosis. CONCLUSION: Collectively, our work indicates that HDM induces ER stress in airway epithelial cells and that ATF6α and ERp57 play a significant role in the development of cardinal features of allergic airways disease. Inhibition of ER stress responses may provide a potential therapeutic avenue in chronic asthma and sub-epithelial fibrosis associated with loss of lung function.


Assuntos
Apoptose , Brônquios/patologia , Estresse do Retículo Endoplasmático/fisiologia , Células Epiteliais/patologia , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Pyroglyphidae/fisiologia , Fator 6 Ativador da Transcrição/deficiência , Fator 6 Ativador da Transcrição/efeitos dos fármacos , Fator 6 Ativador da Transcrição/genética , Animais , Brônquios/metabolismo , Brônquios/fisiopatologia , Caspase 3/metabolismo , Linhagem Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Humanos , Técnicas In Vitro , Cloreto de Metacolina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Isomerases de Dissulfetos de Proteínas/deficiência , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/genética , Fibrose Pulmonar/metabolismo , RNA Interferente Pequeno/farmacologia
6.
Alcohol ; 46(1): 89-99, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21840159

RESUMO

Chronic ethanol consumption increases the risk of type 2 diabetes mellitus, and ethanol has been reported to cause insulin resistance and, inconsistently, to reduce insulin secretion. The mechanism(s) underlying the reduction of insulin secretion by ethanol is not known. We used ß-cell lines and isolated murine islets to determine the effect of ethanol on insulin content and secretion at low- and high-glucose concentrations, in the presence of KCl, diazoxide, tolbutamide, and regulators of cyclic AMP and protein kinase C (PKC). We also determined the gene expression of insulin; pancreas duodenum homeobox 1; and endoplasmic reticulum (ER) stress markers, such as Chop, ERp57, glucose-regulated protein 78/binding immunoglobulin protein, and inositol 1,4,5-triphosphate receptors. Ethanol reduced insulin secretion by interfering with muscarinic signaling and PKC activation but not the K-ATP channels. In addition, ethanol reduced insulin content and caused ER stress. The deleterious effects of ethanol on ß-cells were prevented by 4-methyl pyrazole, an inhibitor of alcohol dehydrogenase, suggesting that ethanol metabolism is required for these effects.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Etanol/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Animais , Linhagem Celular , Chaperona BiP do Retículo Endoplasmático , Expressão Gênica/efeitos dos fármacos , Glucose/genética , Glucose/metabolismo , Proteínas de Choque Térmico/efeitos dos fármacos , Proteínas de Choque Térmico/genética , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/genética , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/genética , Proteína Quinase C/metabolismo , Fator de Transcrição CHOP/efeitos dos fármacos , Fator de Transcrição CHOP/genética
7.
J Neurochem ; 106(1): 333-46, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18384645

RESUMO

In Parkinson's disease, oxidative stress is implicated in protein misfolding and aggregation, which may activate the unfolded protein response by the endoplasmic reticulum (ER). Dopamine (DA) can initiate oxidative stress via H(2)O(2) formation by DA metabolism and by oxidation into DA quinone. We have previously shown that DA quinone induces oxidative protein modification, mitochondrial dysfunction in vitro, and dopaminergic cell toxicity in vivo and in vitro. In this study, we used cysteine- and lysine-reactive fluorescent dyes with 2D difference in-gel electrophoresis, mass spectrometry, and peptide mass fingerprint analysis to identify proteins in PC12 cell mitochondrial-enriched fractions that were altered in abundance following DA exposure (150 muM, 16 h). Quantitative changes in proteins labeled with fluorescent dyes indicated increases in a subset of proteins after DA exposure: calreticulin, ERp29, ERp99, Grp58, Grp78, Grp94 and Orp150 (149-260%), and decreased levels of aldolase A (39-42%). Changes in levels of several proteins detected by 2D difference in-gel electrophoresis were confirmed by western blot. Using this unbiased proteomics approach, our findings demonstrated that in PC12 cells, DA exposure leads to a cellular response indicative of ER stress prior to the onset of cell death, providing a potential link between DA and the unfolded protein response in the pathogenesis of Parkinson's disease.


Assuntos
Dopamina/metabolismo , Retículo Endoplasmático/metabolismo , Frutose-Bifosfato Aldolase/metabolismo , Proteínas de Choque Térmico/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Animais , Calreticulina/efeitos dos fármacos , Calreticulina/metabolismo , Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Eletroforese em Gel Bidimensional , Retículo Endoplasmático/efeitos dos fármacos , Corantes Fluorescentes , Frutose-Bifosfato Aldolase/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/efeitos dos fármacos , Espectrometria de Massas , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/efeitos dos fármacos , Chaperonas Moleculares/metabolismo , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Dobramento de Proteína , Proteínas/efeitos dos fármacos , Proteínas/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
8.
Free Radic Biol Med ; 42(2): 270-9, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17189832

RESUMO

The objectives of this study were to determine the relationships among Type II diabetes (T2DM)-dependent elevations in platelet-derived reactive oxygen species (ROS), platelet-surface protein disulfide isomerase (psPDI) NO-releasing activity, and platelet aggregation and to evaluate the efficacy of rosuvastatin in normalizing these parameters in primary cells derived from a hamster model of prediabetic insulin resistance induced by fructose feeding. Platelets from rosuvastatin-treated non-fructose-fed (NFF) and fructose-fed (FF) hamsters were analyzed for aggregability and psPDI-denitrosation activity. Platelets from NFF animals treated with xanthine/xanthine oxidase (X/XO) were assessed for the same parameters and primary aortic endothelial cells (AEC) cultivated with a range of [rosuvastatin] +/- mevalonate were analyzed for ROS production. Platelets from FF hamsters displayed statistically significant enhanced ROS production, diminished psPDI-mediated NO-releasing activity, and hyperaggregability. Suggestively, platelets from NFF animals treated with X/XO displayed characteristics similar to platelets from FF animals. Rosuvastatin elicited a normalizing effect on all parameters measured in platelets from FF animals. Further, ROS production in primary AEC from FF animals could be blunted to that of NFF animals by concentrations of rosuvastatin in the range of those achieved in the bloodstream. Diminished psPDI-dependent NO-releasing activity and increased initial aggregation rates of FF platelets may result from elevated vascular ROS production under conditions of insulin resistance. Normalization of ROS production and platelet aggregation by rosuvastatin indicates its potential use as a vasculoprotective agent.


Assuntos
Plaquetas/efeitos dos fármacos , Diabetes Mellitus Tipo 2/prevenção & controle , Fluorbenzenos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Estado Pré-Diabético/tratamento farmacológico , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Animais , Antioxidantes , Cricetinae , Frutose/efeitos adversos , Mesocricetus , Óxido Nítrico/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Rosuvastatina Cálcica
10.
J Protein Chem ; 20(2): 155-63, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11563696

RESUMO

The effect of protein aggregates on the aggregation of D-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) during unfolding and refolding has been studied. The aggregation of GAPDH follows a sigmoid course. The presence of protein aggregates increases the aggregation rate during unfolding and refolding of GAPDH but does not change the extent of aggregation and the final renaturation yield. It is suggested that protein aggregates function as seeds for aggregation via hydrophobic interaction with only GAPDH folding intermediates destined to aggregate and do not affect the distribution between pathways leading to correct folding and aggregation. Moreover, two different proteins do not interfere with each other during their simultaneous refolding together in a buffer. These findings provide insight into a mechanism by which cells prevent protein folding against the interference from aggregation of other proteins.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/química , Muramidase/química , Proteínas/química , Animais , Bovinos , Dissulfeto de Glutationa/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/análise , Gliceraldeído-3-Fosfato Desidrogenases/efeitos dos fármacos , Guanidina/farmacologia , Interações Hidrofóbicas e Hidrofílicas , Muramidase/efeitos dos fármacos , Músculos/enzimologia , Parassimpatomiméticos/farmacologia , Fosfatos/farmacologia , Compostos de Potássio/farmacologia , Desnaturação Proteica/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/análise , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Dobramento de Proteína , Coelhos , Soroalbumina Bovina/análise , Soroalbumina Bovina/efeitos dos fármacos , Fatores de Tempo
11.
Biotechniques ; 25(3): 482-8, 490-2, 494, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9762446

RESUMO

Endothelial cell (EC) propagation has been simplified by developing cell-specific selection criteria. Methods commonly used for selectively isolating EC include: (i) differential sieving of disaggregated tissue, (ii) differential plating of cells on extracellular matrices, (iii) lectin affinity isolation of cell populations and (iv) fluorescence-activated cell sorting of cells labeled with a carbocyanine dye of acetylated low-density lipoprotein (DiI-Ac-LDL). Few criteria for selectively propagating pericytes (PC) are currently available. Nonspecific esterases exhibit a high degree of multiplicity when compared with other mammalian isozymes and may be suitable for the identification and selective propagation of cells of the microvasculature. Evaluation of esterase isotype expression in PC and EC by zymography indicates PC contain alpha-naphthyl acetate and alpha-naphthyl butyrate hydrolyzing esterases as well as dipeptidyl peptidase I, while EC only contain alpha-naphthyl acetate esterase. The cytotoxic response of PC and EC to various amino acid esters is assessed by monitoring vital dye uptake and by light microscopy. Several amino acid esters are cytotoxic to both cell types, whereas 50 mM L-leucine methyl ester (L-Leu OMe) is toxic to EC but not to PC. This amino acid ester is also toxic to mesothelial and retinal pigmented epithelial cells, other common contaminants of PC cultures. Analysis of protein composition by two-dimensional gel electrophoresis indicates that L-Leu OMe does not stimulate expression of stress response proteins in PC. Thus, L-Leu OMe can be utilized to cultivate PC selectively from mixed cell populations.


Assuntos
Dipeptídeos/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Imunossupressores/farmacologia , Pericitos/efeitos dos fármacos , Retina/citologia , Aminoácidos/farmacologia , Animais , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/efeitos dos fármacos , Calreticulina , Bovinos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/efeitos dos fármacos , Relação Dose-Resposta a Droga , Esterases/biossíntese , Esterases/efeitos dos fármacos , Ésteres/farmacologia , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/efeitos dos fármacos , Humanos , Pericitos/citologia , Pericitos/enzimologia , Isomerases de Dissulfetos de Proteínas/biossíntese , Isomerases de Dissulfetos de Proteínas/efeitos dos fármacos , Ribonucleoproteínas/biossíntese , Ribonucleoproteínas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...