Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Chem Biodivers ; 20(4): e202201203, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36896496

RESUMO

A novel monoterpene alkaloid, named incarvine G, was isolated from the Incarvillea sinensis Lam. Its chemical structure was elucidated using comprehensive spectroscopic methods. Incarvine G is an ester compound comprised of a monoterpene alkaloid and glucose. This compound showed evident inhibition on cell migration, invasion, and cytoskeleton formation of human MDA-MB-231 with low cytotoxicity.


Assuntos
Antineoplásicos , Bignoniaceae , Monoterpenos , Humanos , Alcaloides/farmacologia , Alcaloides/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Bignoniaceae/química , Estrutura Molecular , Monoterpenos/farmacologia , Monoterpenos/química , Inibição de Migração Celular/efeitos dos fármacos
2.
Bioorg Chem ; 130: 106230, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36375352

RESUMO

Colorectal cancer is a type of cancer encountered worldwide and ranks third among all cancer types in terms of incidence. Polyphenols have been shown to have a wide range of biological functions, including a significant impact on cancer start, development, and promotion through regulating many signaling pathways. The aim of this study was to investigate the anticancer effects of isoeugenol based compounds 1, 2 on HT29 colorectal cancer cell line in vitro. MTT test and scratch assay were carried out to determine the effect of these compounds on HT29 cell proliferation and migration respectively. In addition, mRNA expression levels of apoptosis and metastasis-related genes (p53, Bcl2, Bax, Caspase 3, Caspase7, Caspase8, Caspase9, HIF1-α, VEGF, MMP-2, MMP-9) were examined by quantitative real-time PCR. The results indicated that 1 and 2 inhibited HT29 cell proliferation and induced apoptosis by increasing the Bax/Bcl2 ratio and Caspase-9 and Caspase-3 mRNA expression. In conclusion, the results of this study showed that the treatment of these compounds significantly suppressed the mRNA expressions of metastasis-related genes such as Matrix Metalloproteinase-2, Matrix Metalloproteinase-9, Vascular Endothelial Growth Factor and Hypoxia­Inducible Factor 1α.


Assuntos
Neoplasias do Colo , Metaloproteinase 2 da Matriz , Humanos , Proteína X Associada a bcl-2/metabolismo , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Metaloproteinase 2 da Matriz/genética , Fenóis/química , Fenóis/farmacologia , RNA Mensageiro , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Eugenol/análogos & derivados , Eugenol/química , Eugenol/farmacologia , Inibição de Migração Celular/efeitos dos fármacos
3.
Mol Biol Cell ; 33(1): ar9, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34788129

RESUMO

The ability of cells to sense chemical gradients is essential during development, morphogenesis, and immune responses. Although much is known about chemoattraction, chemorepulsion remains poorly understood. Proliferating Dictyostelium cells secrete a chemorepellent protein called AprA. AprA prevents pseudopod formation at the region of the cell closest to the source of AprA, causing the random movement of cells to be biased away from the AprA. Activation of Ras proteins in a localized sector of a cell cortex helps to induce pseudopod formation, and Ras proteins are needed for AprA chemorepulsion. Here we show that AprA locally inhibits Ras cortical activation through the G protein-coupled receptor GrlH, the G protein subunits Gß and Gα8, Ras protein RasG, protein kinase B, the p21-activated kinase PakD, and the extracellular signal-regulated kinase Erk1. Diffusion calculations and experiments indicate that in a colony of cells, high extracellular concentrations of AprA in the center can globally inhibit Ras activation, while a gradient of AprA that naturally forms at the edge of the colony allows cells to activate Ras at sectors of the cell other than the sector of the cell closest to the center of the colony, effectively inducing both repulsion from the colony and cell differentiation. Together, these results suggest that a pathway that inhibits local Ras activation can mediate chemorepulsion.


Assuntos
Inibição de Migração Celular/fisiologia , Dictyostelium/efeitos dos fármacos , Dictyostelium/metabolismo , Inibição de Migração Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Protozoários/metabolismo , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases Ativadas por p21/metabolismo , Proteínas ras/metabolismo
4.
Sci Rep ; 10(1): 931, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31969633

RESUMO

Wound healing assay is a simple and cost-effective in vitro assay for assessing therapeutic impacts on cell migration. Its key limitation is the possible confoundment by other cellular phenotypes, causing misinterpretation of the experimental outcome. In this study, we attempted to address this problem by developing a simple analytical approach for scoring therapeutic influences on both cell migration and cell death, while normalizing the influence of cell growth using Mitomycin C pre-treatment. By carefully mapping the relationship between cell death and wound closure rate, contribution of cell death and cell migration on the observed wound closure delay can be quantitatively separated at all drug dosing. We showed that both intrinsic cell motility difference and extrinsic factors such as cell seeding density can significantly affect final interpretation of therapeutic impacts on cellular phenotypes. Such discrepancy can be rectified by using the actual wound closure time of each treatment condition for the calculation of phenotypic scores. Finally, we demonstrated a screen for strong pharmaceutical inhibitors of cell migration in cholangiocarcinoma cell lines. Our approach enables accurate scoring of both migrastatic and cytotoxic effects, and can be easily implemented for high-throughput drug screening.


Assuntos
Ensaios de Migração Celular/métodos , Inibição de Migração Celular , Movimento Celular/efeitos dos fármacos , Mitomicina/farmacologia , Cicatrização/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Inibição de Migração Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos
5.
Sci Rep ; 9(1): 7926, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138874

RESUMO

In prostate carcinogenesis, expression and/or activation of the Transient Receptor Potential Melastatin 8 channel (TRPM8) was shown to block in vitro Prostate Cancer (PCa) cell migration. Because of their localization at the plasma membrane, ion channels, such as TRPM8 and other membrane receptors, are promising pharmacological targets. The aim of this study was thus to use nanocarriers encapsulating a TRPM8 agonist to efficiently activate the channel and therefore arrest PCa cell migration. To achieve this goal, the most efficient TRPM8 agonist, WS12, was encapsulated into Lipid NanoCapsules (LNC). The effect of the nanocarriers on channel activity and cellular physiological processes, such as cell viability and migration, were evaluated in vitro and in vivo. These results provide a proof-of-concept support for using TRPM8 channel-targeting nanotechnologies based on LNC to develop more effective methods inhibiting PCa cell migration in zebrafish xenograft.


Assuntos
Anilidas/farmacologia , Inibição de Migração Celular/efeitos dos fármacos , Mentol/análogos & derivados , Neoplasias da Próstata/tratamento farmacológico , Canais de Cátion TRPM/agonistas , Anilidas/administração & dosagem , Humanos , Lipídeos/química , Masculino , Mentol/administração & dosagem , Mentol/farmacologia , Nanocápsulas/química , Células PC-3 , Neoplasias da Próstata/metabolismo , Canais de Cátion TRPM/metabolismo
6.
Toxicol Lett ; 307: 1-10, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-30658152

RESUMO

BACKGROUND: Paraquat (PQ) poisoning is one of the leading causes of suicide attempts in China signature by acute onset of respiratory distress with massive matrix production resulting in progressive pulmonary fibrosis. There is no specific antidote and mortality remains high without effective treatment available. The cellular mechanisms underlying PQ-induced pulmonary fibrosis remain largely unknown. OBJECTIVES: To determine the origin of mesenchymal stem cells (MSCs) migrated to the lung after PQ exposure and their roles in PQ-induced pulmonary fibrosis, to further explore the possible mechanisms involved in these processes, and to help finding novel therapies. METHODS: We used a combination of lineage tracking techniques to investigate the contributions of several cells of MSCs, marked by Nestin or CXCL12, and traced their co-expression of α-smooth muscle actin (α-SMA), a marker for fibrosis, or their co-location with matrix production, marked by collagen-1 production (Col1-GFP) following PQ exposure. Then, we used a CXCL12flox/flox; Prx1-Cre mice and a pharmacologic agent AMD3100 to selectively deplete chemotactic mechanism of the MSCs, and tested pro-fibrotic pathways, fibrotic processes and survival of mice after PQ exposure. RESULTS: Our results showed that after paraquat exposure, the residential Nestin + MSCs were quickly expanded and contributed to extracellular matrix production. Moreover, when we used a CXCL12flox/flox; Prx1-Cre mice to selectively deplete chemotactic mechanism of the MSC, we found that PQ exposure in these mice failed to activate pro-fibrotic pathways including TGF-ß, Wnt and EGFR signaling. Furthermore, when the chemotactic effect of MSCs via CXCL12 was blocked by a pharmacologic agent, AMD3100, it alleviated the development of the fibrotic process and improved survival rate in mice exposed to PQ. CONCLUSION: Collectively, our data suggest paraquat intoxication rapidly activated Nestin + MSCs and that blocking chemotactic effects of MSCs by perivascular CXCL12 inhibition may effectively protect pulmonary injury following paraquat exposure. Our results revealed a novel mechanism for post-PQ lung injury and indicated a novel therapeutic option to attenuate fibrosis induced by paraquat.


Assuntos
Inibição de Migração Celular , Células-Tronco Mesenquimais/efeitos dos fármacos , Paraquat/toxicidade , Fibrose Pulmonar/induzido quimicamente , Animais , Linhagem da Célula , Inibição de Migração Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Feminino , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Nestina/metabolismo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/terapia
7.
Braz. J. Pharm. Sci. (Online) ; 55: e18276, 2019. graf
Artigo em Inglês | LILACS | ID: biblio-1011640

RESUMO

To study the effect of small interfering RNA targeting metastasis-associated lung adenocarcinoma transcript1 (si-MALAT1) combining with curcumin on the invasion and migration abilities of human colon cancer SW480 cells, and to explore the involved molecular mechanism. The recombinant lentiviral vector expressing si-MALAT1 was constructed, and its titer was determined by gradient dilution method. The colon cancer SW480 cells with stable expression of si-MALAT1 was established, followed by treatment with curcumin at different concentrations. The effect of curcumin or si-MALAT1 alone and the combination of the two on the cell activity was detected by MTT assay. The cell invasion and migration abilities were detected by transwell and scratch-wound assay. The relative expression level of MALAT1 was detected by RT-qPCR. The protein expression was determined by Western blot analysis. The IC50 of curcumin alone was 77.69 mmol/L, which was 51.17 mol/L when combined with curcumin and random sequence. The IC50 of curcumin was 30.02 mmol/L when combined with si-MALAT1. The increased susceptibility multiples was 2.58. The wound healing rates were 30.9% and 67.5% after treatment with si-MALAT1 combined with curcumin for 24 hrs and 48 hrs, respectively. The numbers of invasion cells were 200±12, 162±13, 66±8, 53±4 and 16±3 after treatment with si-MALAT1 combined with curcumin for 48 hrs. The relative expression level of lncRNA-MALAT1 in the curcumin group was 68%, and the relative expression level of lncRNA-MALAT1 in si-MALAT1group was 56%, and that for the combination treatment group was about 21%. The protein expression levels of β- catenin, c-myc and cyclinD1 were significantly down-regulated upon treatment with certain concentration of si-MALAT1 alone or combined with curcumin.si-MALAT1 could significantly inhibit the invasion and migration of SW480 cells by enhancing the sensitivity of SW480 cells to curcumin. The mechanism involved mignt be related to the down-regulation of β-catenin, c-myc and cyclinD1 proteins.


Assuntos
Inibição de Migração Celular/efeitos dos fármacos , Neoplasias do Colo , Curcumina/farmacologia , Neoplasias/prevenção & controle , RNA , RNA Interferente Pequeno/efeitos dos fármacos
8.
Am J Med Sci ; 356(4): 374-381, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30360806

RESUMO

BACKGROUND: Cigarette smoking disturbs plasma lipid level and lipoprotein metabolism; however, the effects of smoking on the functional state of high density lipoprotein (HDL) are still not clear. This study aimed to determine the antioxidant and antichemotactic properties of HDL and HDL-mediated cholesterol efflux in healthy subjects after cigarette smoking. MATERIALS AND METHODS: Healthy male subjects, including nonsmokers (n = 16) and chronic smokers (n = 8), were enrolled. After smoking 8 cigarettes within 2 hours, plasma HDL was isolated and tested. Copper-induced low density lipoprotein (LDL) oxidation was used to determine the antioxidant ability of HDL. The concentration of serum amyloid A was measured by Enzyme Linked Immunosorbent Assay. Chemotaxis was detected by transwell assay. HDL-mediated cholesterol efflux was measured using fluorescent cholesterol analog. RESULTS: LDL baseline oxidation state was higher in chronic smokers than that in nonsmokers. Meanwhile, HDL-induced cholesterol efflux in macrophages in chronic smokers was significantly enhanced compared with that in nonsmokers. After acute smoking, both the antioxidant and antichemotactic ability of HDL declined in nonsmokers. However, in healthy chronic smokers, the effect of HDL on the susceptibility of LDL to oxidation was compensatorily enhanced. Nevertheless, their bodies were still in a higher oxidation state. Also, acute smoking did not affect HDL-mediated cholesterol efflux significantly in both nonsmokers and chronic smokers. CONCLUSIONS: Our data suggest that acute smoking attenuates the antioxidant and antichemotactic abilities of HDL in nonsmokers. Chronic smokers are in a higher oxidative state, although the antioxidant function of their HDL is compensatorily enhanced.


Assuntos
Antioxidantes/metabolismo , Inibição de Migração Celular/efeitos dos fármacos , HDL-Colesterol/sangue , Fumar Cigarros/efeitos adversos , Lipoproteínas HDL/sangue , Adulto , Humanos , Masculino , Adulto Jovem
9.
Biochemistry ; 57(28): 4197-4205, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29897736

RESUMO

The autoimmune disease multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), is characterized by an ascending paralysis that is characterized by extensive infiltration of the central nervous system by inflammatory cells. Although several studies to some extent uncover the cellular mechanisms of microglia that govern EAE pathogenesis, the molecular mechanisms that orchestrate the movement of microglia remain unknown, and potential novel therapeutic strategies are still required. In this study, we report that dexmedetomidine, an alpha 2a adrenergic receptor agonist, attenuates the clinical severity of EAE with less infiltration of microglia. During EAE, dexmedetomidine inhibits SDF-1- and I-TAC-induced chemotaxis of microglia mediated by CXCR7 but not CXCR4 or CXCR3. Most importantly, the alpha 2a adrenergic receptor is essential in dexmedetomidine-induced CXCR7 desensitization in microglia. Further experiments confirmed that CXCR7 desensitization required atypical protein kinase C ζ activation, while conventional and novel protein kinase C isoforms were not involved. Altogether, our data elucidate the mechanism of dexmedetomidine-induced CXCR7 desensitization in microglia and amelioration in EAE, which might lead to a better understanding of the therapeutic effects of dexmedetomidine as well as its implications for CXCR7 desensitization in autoimmune disease.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Inibição de Migração Celular/efeitos dos fármacos , Dexmedetomidina/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Microglia/efeitos dos fármacos , Receptores CXCR/imunologia , Animais , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Receptores CXCR/análise
10.
Anticancer Drugs ; 29(4): 341-352, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29420337

RESUMO

MTH1 has become a new rising star in the field of 'cancer phenotypic lethality' and can be targeted in many kinds of tumors. This study aimed to explore the anticancer effect of MTH1-targeted drug (S)-crizotinib on osteosarcoma (OS) cells. We detected MTH1 expression in OS tissues and cells using immunohistochemistry and western blot. The effects of MTH1 on OS cell viability were explored using the siRNA technique and CCK8. The anticancer effects of the MTH1-targeted drug (S)-crizotinib on OS cells were explored by in-vitro assays. The intracellular 8-oxo-dGTP level and oxygen reactive species (ROS) of OS cells were detected by Cy3-conjugated avidin staining and dichlorofluorescein diacetate staining, respectively. The expression of MTH1 was significantly higher in OS tissues and cell lines than that in the corresponding adjacent tissues and osteoblastic cell line. The proliferation of OS cells was significantly inhibited through knockdown of MTH1 by siRNA technology. (S)-Crizotinib could inhibit the proliferation of OS cells with an increase in the apoptosis levels and causing G0/G1 arrest by targeting MTH1 and activating ROS. In addition, (S)-crizotinib could inhibit the migration of OS cells. (S)-Crizotinib could suppress the proliferation and migration, cause G0/G1 arrest, and increase the apoptosis level of OS cells by targeting MTH1 and activating ROS. This study will provide a promising therapeutic target and the theoretical basis for the clinical application of (S)-crizotinib in OS.


Assuntos
Antineoplásicos/farmacologia , Crizotinibe/farmacologia , Enzimas Reparadoras do DNA/biossíntese , Osteossarcoma/tratamento farmacológico , Monoéster Fosfórico Hidrolases/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Adolescente , Adulto , Idoso , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibição de Migração Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Criança , Crizotinibe/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Osteossarcoma/metabolismo , Adulto Jovem
11.
Sci Rep ; 7(1): 15038, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118379

RESUMO

CCR4 is highly expressed on Th2 cells. CCR4 ligands include CCL22 and CCL17. Chemokine-like factor 1 can also mediate chemotaxis via CCR4. We designed and synthetized novel CCR4 antagonists, which were piperazinyl pyridine derivatives, for disrupting the interaction between three ligands and CCR4. We also determined whether these novel CCR4 antagonists could alleviate allergic asthma in a mouse. For identifying the potent compounds in vitro, we used chemotaxis inhibition and competition binding assays induced by CCL22, CCL17 and one of CKLF1's C-terminal peptides, C27. We found compound 8a which showed excellent potency in blocking the interaction of CCR4 and its three ligands. For studying the specificity of compounds, we chose chemotaxis inhibition assays with different receptors and ligands. We found compound 8a had excellent receptor specificity and exerted few influence on the interaction of other receptors and their ligands. In the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, compound 8a had no obvious cytotoxicity till the higher concentration (16 µM). For determining the potency of compounds in blocking the interaction of CCR4 in vivo, we used the ovalbumin induced allergic asthma model in mice. Our study demonstrated that CCR4 blockaded by compound 8a effectively attenuated airway hyperresponsiveness, airway eosinophilia and Th2 cytokines.


Assuntos
Asma/prevenção & controle , Piperazina/química , Pneumonia/prevenção & controle , Piridinas/farmacologia , Receptores CCR4/antagonistas & inibidores , Animais , Asma/metabolismo , Asma/fisiopatologia , Inibição de Migração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células HEK293 , Humanos , Ligantes , Masculino , Camundongos Endogâmicos BALB C , Estrutura Molecular , Pneumonia/metabolismo , Pneumonia/fisiopatologia , Piridinas/química , Receptores CCR4/metabolismo
12.
Eur J Pharm Biopharm ; 119: 310-321, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28694161

RESUMO

CXCR4 and its ligand CXCL12 play a critical role in the metastasis of various types of cancer including breast cancer. Breast tumors preferentially metastasize to the lung, bones and distant lymph nodes, secreting high levels of CXCL12. We hypothesized that targeted inhibition of CXCR4 in breast cancer cells should suppress CXCR4-positive tumor cells toward secondary metastatic sites. In the present study, the efficacy of CXCR4 targeted dendrimers carrying DOX (LFC131-DOX-D4) on cellular binding, cytotoxicity, and migration of BT-549-Luc and T47D breast cancer cells was investigated. PAMAM dendrimers encapsulating DOX was surface functionalized with LFC131 peptide which recognized CXCR4 expressed on the surface of breast cancer cells. The LFC131-DOX-D4 bound to breast cancer cells resulting in significantly enhanced in vitro cellular toxicity as compared with non-targeted dendrimers. The LFC131-D4 exhibited remarkable reduced migration of BT-549-Luc breast cancer cells toward chemoattractant. This report demonstrated the potential utility of LFC131-dendrimer conjugates for breast cancer therapy and metastasis.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibição de Migração Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Dendrímeros/farmacologia , Receptores CXCR4/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Transdução de Sinais/efeitos dos fármacos
13.
J Oral Pathol Med ; 46(7): 484-488, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27732737

RESUMO

BACKGROUND: Oral squamous cell carcinoma is extremely invasive, and this behavior is regulated by binding of extracellular molecules to the cell membrane receptors. The TKI-258 inhibits phosphorylation of FGFRs VEGFRs and PDGFRs. Our aim was to analyze the effect of TKI-258 treatment in cell movement using SCC-4 cell line from human oral squamous cell carcinoma. METHODS: F-actin was stained with rhodamine phalloidin, and confocal analysis was performed. The migration and invasion (membrane covered with Matrigel™ ) three-dimensional assays were performed, and control and cells treated with TKI-258 that migrated through the membrane were counted after 24 h. RESULTS: Control cells presented abundant cytoplasm with F-actin wide distributed and evident cell cortex; however, treated (1, 5 and 10 µM TKI-258) cells showed round morphology, scanty cytoplasm, F-actin disorganized and preserved cell cortex. TKI-258 (1, 5, and 10 µM) treatment inhibits migrating cells (ANOVA, F = 97.749, d.f. = 3, 10; P < 0.0001), and it was concentration dependent. Invading cell treated with 5 µM TKI-258 was significantly lower (t = 6.708, d.f. = 5, P < 0.001). CONCLUSIONS: These results suggest that the tyrosine kinase inhibitor TKI-258 has an inhibitory effect on cell motility, affecting F-actin, cell migration, and cell invasion, and probably, these processes are regulated by signaling pathways FGFRs and/or PDGFRs and/or VEGFRs.


Assuntos
Benzimidazóis/farmacologia , Carcinoma de Células Escamosas/patologia , Inibição de Migração Celular/efeitos dos fármacos , Neoplasias Bucais/patologia , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Actinas/metabolismo , Carcinoma de Células Escamosas/enzimologia , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Neoplasias Bucais/enzimologia , Fosforilação/efeitos dos fármacos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Thromb Res ; 147: 72-79, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27693845

RESUMO

BACKGROUND: Thrombomodulin (TM) alfa, a recombinant human soluble TM, enhances activation of pro-carboxypeptidase B2 (pro-CPB2) by thrombin. Activated pro-CPB2 (CPB2) exerts anti-inflammatory and anti-fibrinolytic activities. Therefore, TM alfa may also have anti-inflammatory and anti-fibrinolytic effects through CPB2. However, these effects of TM alfa have not been elucidated. In the present study, we investigated the effects of TM alfa on inactivation of complement component C5a as an anti-inflammatory effect and prolongation of clot lysis time as an anti-fibrinolytic effect via CPB2 in vitro. METHODS: CPB2 activity and tissue factor-induced thrombin generation was examined by a chromogenic assay. C5a inactivation was evaluated by C-terminal cleavage of C5a and inhibition of C5a-induced human neutrophil migration. Clot lysis time prolongation was examined by a tissue-type plasminogen activator-induced clot lysis assay. RESULTS: CPB2 activity in human plasma was increased by TM alfa and thrombin in a concentration-dependent manner. TM alfa inhibited tissue factor-induced thrombin generation and enhanced pro-CPB2 activation in human plasma simultaneously. The mass spectrum of C5a treated with TM alfa, thrombin, and pro-CPB2 was decreased at 156m/z, indicating that TM alfa enhanced the processing of C5a to C-terminal-cleaved C5a, an inactive form of C5a. C5a-induced human neutrophil migration was decreased after C5a treatment with TM alfa, thrombin, and pro-CPB2. TM alfa prolonged the clot lysis time in human plasma, and this effect was completely abolished by addition of a CPB2 inhibitor. CONCLUSIONS: TM alfa exerts anti-inflammatory and anti-fibrinolytic effects through CPB2 in the presence of thrombin in vitro.


Assuntos
Anti-Inflamatórios/farmacologia , Carboxipeptidase B2/imunologia , Fibrinolíticos/farmacologia , Trombina/antagonistas & inibidores , Ensaios de Migração de Leucócitos , Inibição de Migração Celular/efeitos dos fármacos , Complemento C5a/imunologia , Ativação Enzimática/efeitos dos fármacos , Tempo de Lise do Coágulo de Fibrina , Humanos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Proteínas Recombinantes/farmacologia , Trombina/imunologia , Trombomodulina
15.
Sci Rep ; 6: 35789, 2016 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-27767059

RESUMO

A UPLC-PDA-QTOFMS-guided isolation strategy was employed to screen and track potentially new compounds from Garcinia oblongifolia. As a result, two new prenylated xanthones, oblongixanthones D and E (1-2), six new prenylated benzoylphloroglucinol derivatives, oblongifolins V-Z (3-7) and oblongifolin AA (8), as well as a known compound oblongifolin L (9), were isolated from the EtOAc-soluble fraction of an acetone extract of the leaves of Garcinia oblongifolia guided by UPLC-PDA-QTOFMS analysis. The structures of the new compounds were elucidated by 1D- and 2D-NMR spectroscopic analysis and mass spectrometry. Experimental and calculated ECD spectra were used to determine the absolute configurations. The results of wound healing and transwell migration assay showed that oblongixanthones D (1), E (2), and oblongifolin L (9) have the ability to inhibit cancer cell migration in lower cytotoxic concentrations. Western blotting results showed that these compounds exhibited an anti-metastasis effect mainly through downregulating RAF protein levels. In addition, 2 and 9 could inhibit phospho-MEK and phospho-ERK at downstream. Moreover, 1, 2, and 9 could inhibit snail protein level, suggesting that they could regulate the EMT pathway.


Assuntos
Antineoplásicos Fitogênicos/isolamento & purificação , Inibição de Migração Celular/efeitos dos fármacos , Garcinia/química , Floroglucinol/análogos & derivados , Xantonas/isolamento & purificação , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Ensaios de Seleção de Medicamentos Antitumorais , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células Hep G2 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Floroglucinol/química , Floroglucinol/isolamento & purificação , Folhas de Planta/química , Prenilação , Espectrometria de Massas por Ionização por Electrospray , Xantonas/química , Xantonas/farmacologia
16.
J Immunol ; 196(8): 3429-37, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26969756

RESUMO

Resolvin D1 (RvD1) was shown to be a potent anti-inflammatory and proresolution lipid mediator in several animal models of inflammation, but its mechanism of action in humans is not clear. We show that the RvD1 receptor GPR32 is present on resting, proinflammatory M(LPS) and alternatively activated primary human M(IL-4) macrophages, whereas TGF-ß and IL-6 reduce its membrane expression. Accordingly, stimulation of resting primary human macrophages with 10 nM RvD1 for 48 h maximally reduced the secretion of the proinflammatory cytokines IL-1ß and IL-8; abolished chemotaxis to several chemoattractants like chemerin, fMLF, and MCP-1; and doubled the phagocytic activity of these macrophages toward microbial particles. In contrast, these functional changes were not accompanied by surface expression of markers specific for alternatively activated M(IL-4) macrophages. Similar proresolution effects of RvD1 were observed when proinflammatory M(LPS) macrophages were treated with RvD1. In addition, we show that these RvD1-mediated effects are GPR32 dependent because reduction of GPR32 expression by small interfering RNA, TGF-ß, and IL-6 treatment ablated these proresolution effects in primary human macrophages. Taken together, our results indicate that in humans RvD1 triggers GPR32 to polarize and repolarize macrophages toward a proresolution phenotype, supporting the role of this mediator in the resolution of inflammation in humans.


Assuntos
Anti-Inflamatórios/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Inflamação/tratamento farmacológico , Macrófagos/imunologia , Receptores Acoplados a Proteínas G/imunologia , Inibição de Migração Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Humanos , Inflamação/imunologia , Interleucina-1beta/biossíntese , Interleucina-6/imunologia , Interleucina-8/biossíntese , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Fenótipo , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Fator de Crescimento Transformador beta/imunologia
17.
Arch Toxicol ; 90(7): 1685-94, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26314263

RESUMO

Directional migration of cells to specific locations is required in tissue development, wound healing, and immune responses. Immune cell migration plays a crucial role in both innate and adaptive immunity. Chemokines are small pro-inflammatory chemoattractants that control the migration of leukocytes. In addition, they are also involved in other immune processes such as lymphocyte development and immune pathology. In a previous toxicogenomics study using the Jurkat T cell line, we have shown that the model immunotoxicant TBTO inhibited chemotaxis toward the chemokine CXCL12. In the present work, we aimed at assessing a novel approach to detecting chemicals that affect the process of cell migration. For this, we first evaluated the effects of 31 chemicals on mRNA expression of genes that are known to be related to cell migration. With this analysis, seven immunotoxicants were identified as potential chemotaxis modulators, of which five (CoCl2 80 µM, MeHg 1 µM, ochratoxin A 10 µM, S9-treated ochratoxin A 10 µM, and TBTO 100 nM) were confirmed as chemotaxis inhibitor in an in vitro trans-well chemotaxis assay using the chemokine CXCL12. The transcriptome data of the five compounds together with previously obtained protein phosphorylation profiles for two out of five compounds (i.e., ochratoxin A and TBTO) revealed that the mechanisms behind the chemotaxis inhibition are different for these immunotoxicants. Moreover, the mTOR inhibitor rapamycin had no effect on the chemotaxis of Jurkat cells, indicating that the mTOR pathway is not involved in CXCL12-mediated chemotaxis of Jurkat cells, which is opposite to the findings on human primary T cells (Munk et al. in PLoS One 6(9):e24667, 2011). Thus, the results obtained from the chemotaxis assay conducted with Jurkat cells might not fully represent the results obtained with human primary T cells. Despite this difference, the present study indicated that some compounds may exert their immunotoxic effects through inhibition of CXCL12-mediated chemotaxis.


Assuntos
Inibição de Migração Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Imunossupressores/toxicidade , Linfócitos T/efeitos dos fármacos , Compostos de Trialquitina/toxicidade , Inibição de Migração Celular/genética , Quimiocina CXCL12/imunologia , Quimiotaxia/genética , Humanos , Células Jurkat , RNA Mensageiro/genética , Linfócitos T/imunologia , Transcriptoma/efeitos dos fármacos
18.
Biochem Cell Biol ; 93(6): 604-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26529233

RESUMO

There are a number of reports demonstrating a relationship between the alterations in DFF40 expression and development of some cancers. Here, increased DFF40 expression in T-47D cells in the presence of doxorubicin was envisaged for therapeutic usage. The T-47D cells were transfected with an eukaryotic expression vector encoding the DFF40 cDNA. Following incubation with doxorubicin, propidium iodide (PI) staining was used for cell cycle distribution analysis. The rates of apoptosis were determined by annexin V/PI staining. Apoptosis was also evaluated using the DNA laddering analysis. The viability of DFF40-transfected cells incubated with doxorubicin was significantly decreased compared with control cells. However, there were no substantial changes in the cell cycle distribution of pIRES2-DFF40 cells incubated with doxorubicin compared to control cells. The expression of DFF40, without doxorubicin incubation, had also no significant effect on the cell cycle distribution. There was no DNA laddering in cells transfected with the empty pIRES2 vector when incubated with doxorubicin. In contrast, DNA laddering was observed in DFF40 transfected cells in the presence of doxorubicin after 48 h. Also, the expression of DFF40 and DFF45 was increased in DFF40 transfected cells in the presence of doxorubicin enhancing cell death. Collectively our results indicated that co-treatment of DFF40-transfected cells with doxorubicin can enhance the killing of these tumor cells via apoptosis. Thus, modulation of DFF40 level may be a beneficial strategy for treatment of chemo-resistant cancers.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Desoxirribonucleases/metabolismo , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas de Neoplasias/metabolismo , Inibidores da Topoisomerase II/farmacologia , Antibióticos Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3/genética , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibição de Migração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desoxirribonucleases/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo
19.
Mar Drugs ; 13(10): 6274-90, 2015 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-26473885

RESUMO

A new bioactive macrolactone, nuiapolide (1) was identified from a marine cyanobacterium collected off the coast of Niihau, near Lehua Rock. The natural product exhibits anti-chemotactic activity at concentrations as low as 1.3 µM against Jurkat cells, cancerous T lymphocytes, and induces a G2/M phase cell cycle shift. Structural characterization of the natural product revealed the compound to be a 40-membered macrolactone with nine hydroxyl functional groups and a rare tert-butyl carbinol residue.


Assuntos
Quimiotaxia/efeitos dos fármacos , Cianobactérias/química , Macrolídeos/farmacologia , Divisão Celular/efeitos dos fármacos , Inibição de Migração Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Havaí , Humanos , Células Jurkat , Leucemia de Células T/tratamento farmacológico , Macrolídeos/química , Macrolídeos/isolamento & purificação
20.
J Biol Chem ; 290(35): 21292-304, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26183778

RESUMO

The ELR(-)CXC chemokine CXCL9 is characterized by a long, highly positively charged COOH-terminal region, absent in most other chemokines. Several natural leukocyte- and fibroblast-derived COOH-terminally truncated CXCL9 forms missing up to 30 amino acids were identified. To investigate the role of the COOH-terminal region of CXCL9, several COOH-terminal peptides were chemically synthesized. These peptides display high affinity for glycosaminoglycans (GAGs) and compete with functional intact chemokines for GAG binding, the longest peptide (CXCL9(74-103)) being the most potent. The COOH-terminal peptide CXCL9(74-103) does not signal through or act as an antagonist for CXCR3, the G protein-coupled CXCL9 receptor, and does not influence neutrophil chemotactic activity of CXCL8 in vitro. Based on the GAG binding data, an anti-inflammatory role for CXCL9(74-103) was further evidenced in vivo. Simultaneous intravenous injection of CXCL9(74-103) with CXCL8 injection in the joint diminished CXCL8-induced neutrophil extravasation. Analogously, monosodium urate crystal-induced neutrophil migration to the tibiofemural articulation, a murine model of gout, is highly reduced by intravenous injection of CXCL9(74-103). These data show that chemokine-derived peptides with high affinity for GAGs may be used as anti-inflammatory peptides; by competing with active chemokines for binding and immobilization on GAGs, these peptides may lower chemokine presentation on the endothelium and disrupt the generation of a chemokine gradient, thereby preventing a chemokine from properly performing its chemotactic function. The CXCL9 peptide may serve as a lead molecule for further development of inhibitors of inflammation based on interference with chemokine-GAG interactions.


Assuntos
Anti-Inflamatórios/uso terapêutico , Quimiocina CXCL9/uso terapêutico , Gota/tratamento farmacológico , Interleucina-8/antagonistas & inibidores , Neutrófilos/efeitos dos fármacos , Peptídeos/uso terapêutico , Sequência de Aminoácidos , Animais , Anti-Inflamatórios/química , Inibição de Migração Celular/efeitos dos fármacos , Quimiocina CXCL9/química , Quimiotaxia de Leucócito/efeitos dos fármacos , Glicosaminoglicanos/imunologia , Gota/induzido quimicamente , Gota/imunologia , Humanos , Interleucina-8/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neutrófilos/citologia , Neutrófilos/imunologia , Peptídeos/química , Ácido Úrico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA