Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Mol Pharm ; 14(10): 3436-3447, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28880093

RESUMO

Transporters at the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) play a pivotal role as gatekeepers for efflux or uptake of endogenous and exogenous molecules. The protein expression of a number of them has already been determined in the brains of rodents, nonhuman primates, and humans using quantitative targeted absolute proteomics (QTAP). The dog is an important animal model for drug discovery and development, especially for safety evaluations. The purpose of the present study was to clarify the relevance of the transporter protein expression for drug distribution in the dog brain and CSF. We used QTAP to examine the protein expression of 17 selected transporters and receptors at the dog BBB and BCSFB. For the first time, we directly linked the expression of two efflux transporters, P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), to regional brain and CSF distribution using specific substrates. Two cocktails, each containing one P-gp substrate (quinidine or apafant) and one BCRP substrate (dantrolene or daidzein) were infused intravenously prior to collection of the brain. Transporter expression varied only slightly between the capillaries of different brain regions and did not result in region-specific distribution of the investigated substrates. There were, however, distinct differences between brain capillaries and choroid plexus. Largest differences were observed for BCRP and P-gp: both were highly expressed in brain capillaries, but no BCRP and only low amounts of P-gp were detected in the choroid plexus. Kp,uu,brain and Kp,uu,CSF of both P-gp substrates were indicative of drug efflux. Also, Kp,uu,brain for the BCRP substrates was low. In contrast, Kp,uu,CSF for both BCRP substrates was close to unity, resulting in Kp,uu,CSF/Kp,uu,brain ratios of 7 and 8, respectively. We conclude that the drug transporter expression profiles differ between the BBB and BCSFB in dogs, that there are species differences in the expression profiles, and that CSF is not a suitable surrogate for unbound brain concentrations of BCRP substrates in dogs.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Encéfalo/irrigação sanguínea , Capilares/metabolismo , Plexo Corióideo/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/sangue , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/líquido cefalorraquidiano , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/sangue , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/líquido cefalorraquidiano , Animais , Azepinas/farmacocinética , Transporte Biológico , Barreira Hematoencefálica , Encéfalo/metabolismo , Dantroleno/farmacocinética , Cães , Feminino , Perfilação da Expressão Gênica , Isoflavonas/farmacocinética , Masculino , Proteômica/métodos , Quinidina/farmacocinética , Distribuição Tecidual , Triazóis/farmacocinética
3.
Regul Toxicol Pharmacol ; 88: 238-251, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28634147

RESUMO

With the aim of reconsidering ICH S7B and E14 guidelines, a new in vitro assay system has been subjected to worldwide validation to establish a better prediction platform for potential drug-induced QT prolongation and the consequent TdP in clinical practice. In Japan, CSAHi HEART team has been working on hiPS-CMs in the MEA (hiPS-CMs/MEA) under a standardized protocol and found no inter-facility or lot-to-lot variability for proarrhythmic risk assessment of 7 reference compounds. In this study, we evaluated the responses of hiPS-CMs/MEA to another 31 reference compounds associated with cardiac toxicities, and gene expression to further clarify the electrophysiological characteristics over the course of culture period. The hiPS-CMs/MEA assay accurately predicted reference compounds potential for arrhythmogenesis, and yielded results that showed better correlation with target concentrations of QTc prolongation or TdP in clinical setting than other current in vitro and in vivo assays. Gene expression analyses revealed consistent profiles in all samples within and among the testing facilities. This report would provide CiPA with informative guidance on the use of the hiPS-CMs/MEA assay, and promote the establishment of a new paradigm, beyond conventional in vitro and in vivo assays for cardiac safety assessment of new drugs.


Assuntos
Síndrome do QT Longo/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Cardiotônicos/toxicidade , Eletrodos , Expressão Gênica , Guias como Assunto , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Ativação do Canal Iônico/genética , Japão , Contração Miocárdica/genética , Miócitos Cardíacos/fisiologia
4.
Int Immunol ; 29(3): 133-143, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28338984

RESUMO

IL-17 is known to be a cytokine mainly secreted from Th17 cells, which well associate with autoimmune inflammatory responses. In the generation of Th17 cells, RORc and RORa have pivotal roles in controlling the transcription of Il17. We speculated additional regulation in Il17a transcription and randomly screened a 6344 clone cDNA library to identify specific modulators for Il17a promoter activity. After the screen, the E3 ubiquitin ligases SIAH1 and SIAH2 were investigated further and confirmed to increase Il17a promoter activity in a T-cell line and to promote Th17 development ex vivo. This enhancement was a consequence of enhanced expression of hypoxia-inducible factor-1α (HIF-1α) protein, which is reported to directly regulate expression of Il17a and Rorgt at the transcriptional level. In the absence of HIF-1α, both ubiquitin ligases had little effect on Th17 cell differentiation. These results suggest that the SIAH1 and SIAH2 play a pivotal role to promote Th17 cell differentiation through maintaining the stability of HIF-1α protein.


Assuntos
Diferenciação Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Th17/citologia , Células Th17/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ubiquitina-Proteína Ligases/genética
5.
Mol Imaging Biol ; 19(5): 746-753, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28050748

RESUMO

PURPOSE: In vivo detection of pathological insults during the early stages of rheumatoid synovitis is essential to allow early anti-inflammatory treatment for prevention of joint destruction. Whether rheumatoid synovitis pathology and the efficacy of therapies can be visualized by positron emission tomography (PET) tracers specific to the inflammatory process was investigated. PROCEDURES: Using a collagen-induced experimental rat model of rheumatoid arthritis, in vivo imaging using the PET tracers [11C]PK11195, which binds to the translocator protein mainly expressed on myeloid cells, and [11C]ketoprofen, for cyclooxygenase imaging, was performed. To evaluate therapeutic efficacy, model animals were administered the tumour necrosis factor alpha blocker etanercept subcutaneously. RESULTS: [11C]PK11195 and [11C]ketoprofen uptakes were significantly higher in inflamed paws of collagen-induced arthritis rats than in normal rats. The data showed a correlation between tracer uptake values and paw swelling. After treatment with etanercept, [11C]ketoprofen uptake was significantly lower in treated animals than in untreated ones, whereas [11C]PK11195 uptake in the inflamed regions was comparable to that in the untreated group. CONCLUSIONS: With [11C]PK11195 and [11C]ketoprofen tracers, non-invasive in vivo PET imaging for rheumatoid synovitis can provide diagnostic evidence of early synovitis and allow monitoring inflammatory cell activity during treatment.


Assuntos
Artrite Reumatoide/diagnóstico por imagem , Artrite Reumatoide/diagnóstico , Radioisótopos de Carbono/química , Isoquinolinas/química , Cetoprofeno/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Animais , Artrite Reumatoide/patologia , Modelos Animais de Doenças , Feminino , Imageamento Tridimensional , Inflamação/patologia , Articulações/patologia , Ratos Endogâmicos Lew , Tomografia Computadorizada por Raios X , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
6.
Eur J Nutr ; 56(3): 949-964, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26704713

RESUMO

PURPOSE: The rhizome of Kaempferia parviflora (KP) is used in traditional Thai medicine. In this study, we investigated the effects of an ethanol KP extract and two of its components [5,7-dimethoxyflavone (DMF) and 5-hydroxy-3,7,3',4'-tetramethoxyflavone (TMF)] on monocyte adhesion and cellular reactive oxygen species (ROS) production in human umbilical vein endothelial cells (HUVECs), which provide an in vitro model of events relevant to the development and progression of atherosclerosis. METHODS: RAW264.7 mouse macrophage-like cells were incubated with various concentrations of KP extract or polymethoxyflavonoids and stimulated with lipopolysaccharide prior to measuring nitrite levels in the culture media. Monocyte adhesion was evaluated by measuring the fluorescently labeled human monocytic leukemia THP-1 cells that is attached to tumor necrosis factor-α (TNF-α)-stimulated HUVECs. Cellular ROS production was assessed by measuring cellular antioxidant activity using pyocyanin-stimulated HUVECs. RESULTS: KP extract and DMF reduced nitrite levels (as indicator of nitric oxide production) in LPS-stimulated RAW264.7 cells and also inhibited THP-1 cell adhesion to HUVECs. These treatments induced mRNA expression of endothelial nitric oxide synthase in TNF-α-stimulated HUVECs and downregulated that of various cell adhesion molecules, inflammatory mediators, and endothelial function-related genes. Angiotensin-converting enzyme activity was inhibited by KP extract in vitro. Furthermore, KP extract, DMF, and TMF inhibited the production of cellular ROS in pyocyanin-stimulated HUVECs. CONCLUSION: KP extract, DMF, and TMF showed potential anti-inflammatory and antioxidant effects in these in vitro models, properties that would inhibit the development and progression of atherosclerosis.


Assuntos
Adesão Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Zingiberaceae/química , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Regulação para Baixo , Flavonoides/farmacologia , Humanos , Lipopolissacarídeos/metabolismo , Camundongos , Monócitos/citologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-27646297

RESUMO

Recent increasing evidence suggests that the currently-used platforms in vitro IKr and APD, and/or in vivo QT assays are not fully predictive for TdP, and do not address potential arrhythmia (VT and/or VF) induced by diverse mechanisms of action. In addition, other cardiac safety liabilities such as functional dysfunction of excitation-contraction coupling (contractility) and structural damage (morphological damage to cardiomyocytes) are also major causes of drug attrition, but current in vitro assays do not cover all these liabilities. We organized the Consortium for Safety Assessment using Human iPS cells (CSAHi; http://csahi.org/en/), based on the Japan Pharmaceutical Manufacturers Association (JPMA), to verify the application of human iPS/ES cell-derived cardiomyocytes in drug safety evaluation. The main goal of the CSAHi HEART team has been to propose comprehensive screening strategies to predict a diverse range of cardiotoxicities by using recently introduced platforms (multi-electrode array (MEA), patch clamp, cellular impedance, motion field imaging [MFI], and Ca transient systems) while identifying the strengths and weaknesses of each. Our study shows that hiPS-CMs used in these platforms have pharmacological responses more relevant to humans in comparison with existent hERG, APD or Langendorff (MAPD/contraction) assays, and not only MEA but also other methods such as impedance, MFI, and Ca transient systems would offer paradigm changes of platforms for predicting drug-induced QT risk and/or arrhythmia or contractile dysfunctions. Furthermore, we propose a potential multi-parametric platform in which field potential (MEA)-Ca transient-contraction (MFI) could be evaluated simultaneously as an ideal novel platform for predicting a diversity of cardiac toxicities, namely whole effects on the excitation-contraction cascade.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Modelos Cardiovasculares , Miócitos Cardíacos/efeitos dos fármacos , Cardiotoxicidade , Técnicas de Cultura de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Microeletrodos , Miócitos Cardíacos/fisiologia , Preparações Farmacêuticas/administração & dosagem
8.
Regul Toxicol Pharmacol ; 77: 75-86, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26884090

RESUMO

In vitro screening of hERG channels are recommended under ICH S7B guidelines to predict drug-induced QT prolongation and Torsade de Pointes (TdP), whereas proarrhythmia is known to be evoked by blockage of other ion channels involved in cardiac contraction and compensation mechanisms. A consortium for drug safety assessment using human iPS cells-derived cardiomyocytes (hiPS-CMs), CSAHi, has been organized to establish a novel in vitro test system that would enable better prediction of drug-induced proarrhythmia and QT prolongation. Here we report the inter-facility and cells lot-to-lot variability evaluated with FPDc (corrected field potential duration), FPDc10 (10% FPDc change concentration), beat rate and incidence of arrhythmia-like waveform or arrest on hiPS-CMs in a multi-electrode array system. Arrhythmia-like waveforms were evident for all test compounds, other than chromanol 293B, that evoked FPDc prolongation in this system and are reported to induce TdP in clinical practice. There was no apparent cells lot-to-lot variability, while inter-facility variabilities were limited within ranges from 3.9- to 20-folds for FPDc10 and about 10-folds for the minimum concentration inducing arrhythmia-like waveform or arrests. In conclusion, the new assay model reported here would enable accurate prediction of a drug potential for proarrhythmia.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Diferenciação Celular , Canal de Potássio ERG1/antagonistas & inibidores , Frequência Cardíaca/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Microeletrodos , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/toxicidade , Testes de Toxicidade/instrumentação , Potenciais de Ação , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Bioensaio , Cardiotoxicidade , Técnicas de Cultura de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/metabolismo , Desenho de Equipamento , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Japão , Miócitos Cardíacos/metabolismo , Observação , Reprodutibilidade dos Testes , Medição de Risco , Testes de Toxicidade/métodos , Testes de Toxicidade/normas
9.
J Pharm Sci ; 105(2): 876-883, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26429295

RESUMO

Understanding how transporters contribute to the distribution of inhaled drugs in the lung is important for the discovery and development of such drugs. Protein expression levels may be useful to predict and understand drug distribution. As previously reported, organic cation/carnitine transporter 1 (OCTN1) and multidrug resistance-associated protein 1 (MRP1) have higher levels of protein expression among transporters in primary cultured human lung cells. Nevertheless, it is unclear to what extent transport activity correlates with transporter protein expression. The purpose is to evaluate whether differences in OCTN1 and MRP1 protein expression govern the respective transport activity in primary cultured human lung cells. The model substrates of 4-[4-(dimethylamino) styryl]-N-methylpyridinium iodide (ASP(+)) and carboxy-dichlorofluorescein (CDF) for OCTN1 and MRP1, respectively, were used in the lung cells from five donors. Significant correlation was found between the kinetic parameter Vmax for ASP(+) and OCTN1 protein expression in plasma membrane of tracheal, bronchial, and alveolar cells (r(2) = 0.965, 0.834, and 0.877, respectively), and between the efflux of CDF and MRP1 protein expression in plasma membrane of tracheal, bronchial, and alveolar cells (r(2) = 0.800, 0.904, and 0.790, respectively). These findings suggest that OCTN1 and MRP1 protein concentrations are determinants for drug distribution in the lung.


Assuntos
Brônquios/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Traqueia/metabolismo , Brônquios/citologia , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Transporte Proteico/fisiologia , Alvéolos Pulmonares/citologia , Simportadores , Traqueia/citologia
11.
J Pharm Sci ; 104(9): 3029-38, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25690838

RESUMO

Understanding the mechanisms of drug transport in the human lung is an important issue in pulmonary drug discovery and development. For this purpose, there is an increasing interest in immortalized lung cell lines as alternatives to primary cultured lung cells. We recently reported the protein expression in human lung tissues and pulmonary epithelial cells in primary culture, (Sakamoto A, Matsumaru T, Yamamura N, Uchida Y, Tachikawa M, Ohtsuki S, Terasaki T. 2013. J Pharm Sci 102(9):3395-3406) whereas comprehensive quantification of protein expressions in immortalized lung cell lines is sparse. Therefore, the aim of the present study was to clarify the drug transporter protein expression of five commercially available immortalized lung cell lines derived from tracheobronchial cells (Calu-3 and BEAS2-B), bronchiolar-alveolar cells (NCI-H292 and NCI-H441), and alveolar type II cells (A549), by liquid chromatography-tandem mass spectrometry-based approaches. Among transporters detected, breast cancer-resistance protein in Calu-3, NCI-H292, NCI-H441, and A549 and OCTN2 in BEAS2-B showed the highest protein expression. Compared with data from our previous study,(Sakamoto A, Matsumaru T, Yamamura N, Uchida Y, Tachikawa M, Ohtsuki S, Terasaki T. 2013. J Pharm Sci 102(9):3395-3406) NCI-H441 was the most similar with primary lung cells from all regions in terms of protein expression of organic cation/carnitine transporter 1 (OCTN1). In conclusion, the protein expression profiles of transporters in five immortalized lung cell lines were determined, and these findings may contribute to a better understanding of drug transport in immortalized lung cell lines.


Assuntos
Transporte Biológico/fisiologia , Bronquíolos/metabolismo , Células Epiteliais/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Alvéolos Pulmonares/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Humanos , Neoplasias Pulmonares/metabolismo , Espectrometria de Massas em Tandem/métodos
12.
Biochem Biophys Res Commun ; 451(2): 215-21, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25088998

RESUMO

IL-27 is a heterodimeric cytokine that regulates both innate and adaptive immunity. The immunosuppressive effect of IL-27 largely depends on induction of IL-10-producing Tr1 cells. To date, however, effects of IL-27 on regulation of immune responses via mediators other than cytokines remain poorly understood. To address this issue, we examined immunoregulatory effects of conditional medium of bone marrow-derived macrophages (BMDMs) from WSX-1 (IL-27Rα)-deficient mice and found enhanced IFN-γ and IL-17A secretion by CD4(+) T cells as compared with that of control BMDMs. We then found that PGE2 production and COX-2 expression by BMDMs from WSX-1-deficient mice was increased compared to control macrophages in response to LPS. The enhanced production of IFN-γ and IL-17A was abolished by EP2 and EP4 antagonists, demonstrating PGE2 was responsible for enhanced cytokine production. Murine WSX-1-expressing Raw264.7 cells (mWSX-1-Raw264.7) showed phosphorylation of both STAT1 and STAT3 in response to IL-27 and produced less amounts of PGE2 and COX-2 compared to parental RAW264.7 cells. STAT1 knockdown in parental RAW264.7 cells and STAT1-deficiency in BMDMs showed higher COX-2 expression than their respective control cells. Collectively, our result indicated that IL-27/WSX-1 regulated PGE2 secretion via STAT1-COX-2 pathway in macrophages and affected helper T cell response in a PGE2-mediated fashion.


Assuntos
Dinoprostona/biossíntese , Interleucinas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linhagem Celular , Meios de Cultivo Condicionados , Ciclo-Oxigenase 2/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Interferon gama/biossíntese , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Citocinas/deficiência , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de Interleucina , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/metabolismo
13.
Biosci Biotechnol Biochem ; 78(5): 851-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25035989

RESUMO

The rhizome of Kaempferia parviflora has been used in traditional Thai medicine. In this study, we identified and compared specific compounds from the hexane extract of K. parviflora with those from other Zingiberaceous plants by using gas chromatography-mass spectrometry. We identified 5,7-dimethoxyflavone (DMF), 5-hydroxy-3,7,3',4'-tetramethoxyflavone (TMF), estimated 3,5,7-trimethoxyflavone, 5-hydroxy-7,4'-dimethoxyflavone, 3,5,7,4'-tetramethoxyflavone, and investigated their anti-inflammatory effects in rat basophilic leukemia (RBL-2H3) cells stimulated with an IgE antigen or a calcium ionophore. We found that DMF and TMF more potently inhibited antigen-induced degranulation than did nobiletin, a well-known anti-inflammatory agent. In addition, compared to RBL-2H3 cells stimulated with a calcium ionophore, those treated with DMF and TMF showed more marked inhibition of the degranulation and the production and mRNA expression of inflammatory mediators. These results suggest that DMF and TMF inhibit an early step in the high-affinity IgE receptor signaling cascade rather than intracellular calcium release and protein kinase C activation.


Assuntos
Anti-Inflamatórios/análise , Anti-Inflamatórios/farmacologia , Extratos Vegetais/análise , Extratos Vegetais/farmacologia , Zingiberaceae/química , Animais , Anti-Inflamatórios/isolamento & purificação , Degranulação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cromatografia Líquida , Flavonoides/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Hexanos/química , Mediadores da Inflamação/metabolismo , Extratos Vegetais/isolamento & purificação , Ratos
14.
Arterioscler Thromb Vasc Biol ; 33(5): 935-42, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471233

RESUMO

OBJECTIVE: Macrophage (Mϕ) migration rests on the adhesion/detachment between Mϕ surface components and extracellular matrixes, and the contribution of numerous inflammatory disorders. Plasminogen activator inhibitor (PAI)-1, a serine protease inhibitor, influences Mϕ motility through an action distinct from its classical modulation of the plasmin-based fibrinolytic process. We rely here on a small molecule PAI-1 inhibitor (TM5275) to investigate the role of PAI-1 in Mϕ migration in the pathogenesis of renal injury. APPROACH AND RESULTS: Mϕ migration was inhibited both in vitro and in vivo by TM5275. It was also reduced in T-cell-deficient nude mice, but not in PAI-1-deficient mice. Mϕ migration hinged on the interaction of PAI-1 with low-density lipoprotein receptor-related protein, an interaction prevented by TM5275, but not with vitronectin, urokinase-type plasminogen activator, or tissue-type plasminogen activator. Fed to rats with anti-Thy-1-induced nephritis, TM5275 significantly decreased Mϕ accumulation and ameliorated the progression of renal injury. CONCLUSIONS: These findings suggest that a small molecule PAI-1 inhibitor represents a novel class of anti-inflammatory agents targeting Mϕ migration by the inhibition of the interaction of PAI-1 with low-density lipoprotein receptor-related protein.


Assuntos
Macrófagos/efeitos dos fármacos , Piperazinas/farmacologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , para-Aminobenzoatos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Movimento Celular/efeitos dos fármacos , Glomerulonefrite/patologia , Isoanticorpos/farmacologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/fisiologia , Camundongos , Ratos
15.
Biochem Biophys Res Commun ; 431(4): 772-6, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23333329

RESUMO

The CXCL12/CXCR4 axis is involved in many cellular responses for host homeostasis, and malfunction of this signaling pathway is associated with a variety of diseases. It is now known that CXCL12 also binds to another newly identified chemokine receptor, CXCR7, which does not couple with a G-protein. CXCR7 can form homodimers, or heterodimers with CXCR4, and is believed to sequester the chemokine CXCL12, although the CXCL12/CXCR7 axis activates MAP kinases through ß-arrestin. Therefore, it has not been well defined how CXCR7 activation affects CXCL12-induced cellular events. To elucidate the function of CXCR7, we prepared CXCR7 agonist Compound 1. Compound 1 is a selective and potent CXCR7 agonist that clearly has the activity to recruit ß-arrestin toward CXCR7. It also activates MAP kinases Akt and ERK. Using this compound, we confirmed that the CXCR7 agonist, but not an antagonistic antibody, did inhibit CXCL12 induced HUVEC tube formation, suggesting that activation of CXCR7 ameliorates CXCL12 induced cellular events, probably by affecting on CXCR4 function. We show that ß-arrestin recruitment to CXCR4 is reduced by over-expression of CXCR7 and activation of CXCR7 by agonist treatment reduces the protein level of CXCR4. Based on our results, together with reported information, we propose that CXCR7, when up-regulated upon inflammation, can act as a negative regulator of CXCR4 by heterodimerizing with CXCR4, inducing its internalization and degradation. This mechanism suggests that CXCR7 agonists can have a therapeutic effect on CXCL12 causing diseases by countering the effects of CXCL12.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Piridinas/farmacologia , Quinolonas/farmacologia , Receptores CXCR4/metabolismo , Receptores CXCR/agonistas , Regulação para Baixo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Piridinas/química , Quinolonas/química
16.
Int Immunol ; 24(9): 539-50, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22914861

RESUMO

Spleen tyrosine kinase (Syk) is associated with Fcγ receptors (FcγRs) and transmits activation signals through FcγRs in myeloid cells. Thus, application of drugs to inhibit Syk activity can affect the development of immune diseases mediated by autoantibodies, while unexpected systemic effects by the inhibition may be concerned because Syk has multiple physiological functions. We used tamoxifen-inducible systemic conditional Syk knockout (KO) mice to evaluate the role of Syk in the pathogenesis of autoimmune arthritis and to investigate the systemic effects of Syk deletion. In a collagen antibody-induced arthritis model, Syk KO mice were almost completely protected from disease induction and showed significantly attenuated accumulation of neutrophils and macrophages in the joints. Syk-deleted macrophages showed less IL-6 and MCP-1 production upon FcγR ligation and exhibited reduced FcγR-mediated phagocytosis in vitro. Syk-deleted macrophages produce more RANTES upon FcγR ligation, indicating a Syk-independent signaling through the FcγR. We further found that both wild-type and Syk-deleted macrophages induced neutrophil chemotaxis upon FcγR ligation in vitro, and air-pouch model demonstrated that Syk-deleted neutrophils have a potential to infiltrate into local tissues in response to collagen and anti-collagen antibodies. However, Syk-deleted neutrophils exhibited greatly decreased neutrophil extracellular traps formation and FcγR-mediated phagocytosis. Our results indicated that Syk deficiency rendered mice completely unresponsive to immune activation by anti-collagen antibodies with disabling one pathway of FcγR-mediated signaling that was crucial for arthritis induction.


Assuntos
Artrite Experimental/imunologia , Autoanticorpos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/imunologia , Neutrófilos/imunologia , Proteínas Tirosina Quinases/metabolismo , Animais , Autoanticorpos/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Colágeno/imunologia , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Fagocitose , Proteínas Tirosina Quinases/genética , Receptores de IgG/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Quinase Syk
17.
PLoS One ; 7(7): e40343, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792286

RESUMO

Homeostatic regulation of epidermal keratinocytes is controlled by the local cytokine milieu. However, a role for suppressor of cytokine signaling (SOCS), a negative feedback regulator of cytokine networks, in skin homeostasis remains unclear. Keratinocyte specific deletion of Socs3 (Socs3 cKO) caused severe skin inflammation with hyper-production of IgE, epidermal hyperplasia, and S100A8/9 expression, although Socs1 deletion caused no inflammation. The inflamed skin showed constitutive STAT3 activation and up-regulation of IL-6 and IL-20 receptor (IL-20R) related cytokines, IL-19, IL-20 and IL-24. Disease development was rescued by deletion of the Il6 gene, but not by the deletion of Il23, Il4r, or Rag1 genes. The expression of IL-6 in Socs3 cKO keratinocytes increased expression of IL-20R-related cytokines that further facilitated STAT3 hyperactivation, epidermal hyperplasia and neutrophilia. These results demonstrate that skin homeostasis is strictly regulated by the IL-6-STAT3-SOCS3 axis. Moreover, the SOCS3-mediated negative feedback loop in keratinocytes has a critical mechanistic role in the prevention of skin inflammation caused by hyperactivation of STAT3.


Assuntos
Dermatite/metabolismo , Interleucinas/fisiologia , Receptores de Interleucina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Dermatite/imunologia , Dermatite/patologia , Expressão Gênica , Hiperplasia/imunologia , Hiperplasia/metabolismo , Hiperplasia/patologia , Interleucinas/genética , Interleucinas/metabolismo , Queratinócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/fisiologia , beta-Defensinas/genética , beta-Defensinas/metabolismo
18.
J Immunol ; 182(11): 7212-21, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19454718

RESUMO

TLR stimulation triggers a signaling pathway via MyD88 and IL-1R-associated kinase 4 that is essential for proinflammatory cytokine induction. Although NF-kappaB has been shown to be one of the key transcriptional regulators of these cytokines, evidence suggests that other factors may also be important. In this study, we showed that MyD88-deficient macrophages have defective c-Rel activation, which has been linked to IL-12p40 induction, but not IL-6 or TNF-alpha. We also investigated other transcription factors and showed that C/EBPbeta and C/EBPdelta expression was limited in MyD88- or IL-1R-associated kinase 4-deficient macrophages treated with LPS. Importantly, the absence of both C/EBPbeta and C/EBPdelta resulted in the impaired induction of proinflammatory cytokines stimulated by several TLR ligands. Our results identify c-Rel and C/EBPbeta/delta as important transcription factors in a MyD88-dependent pathway that regulate the induction of proinflammatory cytokines.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Proteína delta de Ligação ao Facilitador CCAAT/fisiologia , Citocinas/biossíntese , Proteínas Proto-Oncogênicas c-rel/fisiologia , Receptores Toll-Like/imunologia , Animais , Células Cultivadas , Mediadores da Inflamação , Quinases Associadas a Receptores de Interleucina-1 , Macrófagos , Camundongos , Fator 88 de Diferenciação Mieloide/deficiência , Ativação Transcricional/imunologia
19.
J Endocrinol ; 198(3): 489-97, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18579725

RESUMO

Nuclear receptor subfamily 5, group A, member 1 (NR5A1 previously known as SF-1/AD4BP) is a transcription factor involved in the development of adrenal/gonadal tissues and steroidogenic lineage cell differentiation in adult somatic stem cells. To understand the cellular signaling network that regulates NR5A1 gene expression, loss of function screening with an siRNA kinome library, and gain of function screening with an addressable full-length cDNA library representing one quarter of the human genome was carried out. The NR5A1 gene expression was activated in mesenchymal stem cells by siRNA directed against protein kinase C (PKC)-delta, erb-B3, RhoGAP (ARHGAP26), and hexokinase 2, none of which were previously known to be involved in the NR5A1 gene expression. Among these, we identified crosstalk between erb-B3 and PKC-delta signaling cascades. In addition, the gain of function studies indicated that sex-determining region Y (SRY)-box 15 (SOX15), TEA domain family member 4, KIAA1257 (a gene of unknown function), ADAM metallopeptidase with thrombospondin type 1 motif 6, Josephin domain containing 1, centromere protein, TATA box-binding protein-associated factor 5-like RNA polymerase, and inducible T-cell co-stimulator activate NR5A1 gene expression. These results provide new insights into the molecular mechanisms of NR5A1 gene expression.


Assuntos
Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Adenoviridae/genética , Animais , Bovinos , Linhagem Celular , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , AMP Cíclico/farmacologia , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Biblioteca Gênica , Vetores Genéticos/genética , Hexoquinase/genética , Hexoquinase/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Regiões Promotoras Genéticas/genética , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , RNA Interferente Pequeno/genética , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
J Immunol ; 179(7): 4598-607, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878357

RESUMO

In innate immunity, microbial components stimulate macrophages to produce antimicrobial substances, cytokines, other proinflammatory mediators, and IFNs via TLRs, which trigger signaling pathways activating NF-kappaB, MAPKs, and IFN response factors. We show in this study that, in contrast to its activating role in T cells, in macrophages the protein phosphatase calcineurin negatively regulates NF-kappaB, MAPKs, and IFN response factor activation by inhibiting the TLR-mediated signaling pathways. Evidence for this novel role for calcineurin was provided by the findings that these signaling pathways are activated when calcineurin is inhibited either by the inhibitors cyclosporin A or FK506 or by small interfering RNA-targeting calcineurin, and that activation of these pathways by TLR ligands is inhibited by the overexpression of a constitutively active form of calcineurin. We further found that IkappaB-alpha degradation, MAPK activation, and TNF-alpha production by FK506 were reduced in macrophages from mice deficient in MyD88, Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF), TLR2, or TLR4, whereas macrophages from TLR3-deficient or TLR9 mutant mice showed the same responses to FK506 as those of wild-type cells. Biochemical studies indicate that calcineurin interacts with MyD88, TRIF, TLR2, and TLR4, but not with TLR3 or TLR9. Collectively, these results suggest that calcineurin negatively regulates TLR-mediated activation pathways in macrophages by inhibiting the adaptor proteins MyD88 and TRIF, and a subset of TLRs.


Assuntos
Calcineurina/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Calcineurina/genética , Inibidores de Calcineurina , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptores do Fator de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...