Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Nanomedicine ; 19: 4923-4939, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38828201

RESUMO

Purpose: In recent years, exosomes have been proved to be used to treat many diseases. However, due to the lack of uniform quality control standards for exosomes, the safety of exosomes is still a problem to be solved, especially now more and more exosomes are used in clinical trials, and its non-clinical safety evaluation is particularly important. However, there is no safety evaluation standard for exosomes at present. Therefore, this study will refer to the evaluation criteria of therapeutic biological products, adopt non-human primates to evaluate the non-clinical safety of human umbilical cord mesenchymal stem cell exosomes from the general pharmacology and immunotoxicity, aiming at establishing a safety evaluation system of exosomes and providing reference for the clinical application of exosomes in the future. Methods: 3.85 × 1012 exosomes derived from human umbilical cord mesenchymal stem cells were injected into cynomolgus monkeys intravenously. The changes of general clinical conditions, hematology, immunoglobulin, Th1/Th2 cytokines, T lymphocytes and B lymphocytes, and immune organs were observed before and within 14 days after injection. Results: The results showed that exosomes did not have obvious pathological effects on the general clinical conditions, blood, coagulation function, organ coefficient, immunoglobulin, Th1/Th2 cytokines, lymphocytes, major organs, and major immune organs (spleen, thymus, bone marrow) of cynomolgus monkeys. However, the number of granulocyte-macrophage colonies in exosomes group was significantly higher than that in control group. Conclusion: To sum up, the general pharmacological results and immunotoxicity results showed that the injection of 3.85 × 1012 exosomes may have no obvious adverse reactions to cynomolgus monkeys. This dose of exosomes is relatively safe for treatment, which provides basis research for non-clinical safety evaluation of exosomes and provides reliable research basis for future clinical application of exosomes.


Assuntos
Exossomos , Macaca fascicularis , Células-Tronco Mesenquimais , Cordão Umbilical , Animais , Exossomos/química , Células-Tronco Mesenquimais/citologia , Humanos , Cordão Umbilical/citologia , Masculino , Feminino , Citocinas/metabolismo
2.
Differentiation ; 135: 100744, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38128465

RESUMO

Differentiation of human umbilical cord mesenchymal stem cells (Uc-MSCs) into islet-like clusters which are capable of synthesizing and secreting insulin can potentially serve as donors for islet transplantation in the patient deficiency in islet ß cell function both in type 1 or type 2 diabetic patients. Therefore, we developed an easy and higher efficacy approach by trypsinazing the Uc-MSCs and followed culture in differentiation medium to induce of Uc-MSCs differentiation into islet-like clusters, and the potential mechanism that in the early stage of differentiation was also investigated by using RNA-sequencing and bioinformatics. Results show that induction efficacy was reached to 98% and TGF-ß signaling pathway may play critical role in the early stage differentiation, it was further confirmed that the retardant effect of differentiation progress either in cell morphology or in islet specific genes expression can be observed upon blocking the activation of TGF-ß signaling pathway using specific inhibitor of LY2109761 (TßRI/II kinase inhibitor). Our current study, for the first time, development a protocol for differentiation of Uc-MSCs into islet-like clusters, and revealed the importance of TGF-ß signaling pathway in the early stage of differentiation of Uc-MSCs into islet-like clusters. Our study will provide alternative approach for clinical treatment of either type I or type II diabtes mellitus with dysfunctional pancreatic islets.


Assuntos
Células Secretoras de Insulina , Células-Tronco Mesenquimais , Humanos , Insulina , Tripsina/metabolismo , Diferenciação Celular/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Transdução de Sinais , Cordão Umbilical
3.
PLoS Biol ; 21(6): e3002142, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37289842

RESUMO

Rab26 is known to regulate multiple membrane trafficking events, but its role in insulin secretion in pancreatic ß cells remains unclear despite it was first identified in the pancreas. In this study, we generated Rab26-/- mice through CRISPR/Cas9 technique. Surprisingly, insulin levels in the blood of the Rab26-/- mice do not decrease upon glucose stimulation but conversely increase. Deficiency of Rab26 promotes insulin secretion, which was independently verified by Rab26 knockdown in pancreatic insulinoma cells. Conversely, overexpression of Rab26 suppresses insulin secretion in both insulinoma cell lines and isolated mouse islets. Islets overexpressing Rab26, upon transplantation, also failed to restore glucose homeostasis in type 1 diabetic mice. Immunofluorescence microscopy revealed that overexpression of Rab26 results in clustering of insulin granules. GST-pulldown experiments reveal that Rab26 interacts with synaptotagmin-1 (Syt1) through directly binding to its C2A domain, which interfering with the interaction between Syt1 and SNAP25, and consequently inhibiting the exocytosis of newcomer insulin granules revealed by TIRF microscopy. Our results suggest that Rab26 serves as a negative regulator of insulin secretion, via suppressing insulin granule fusion with plasma membrane through sequestering Syt1.


Assuntos
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Insulinoma , Ilhotas Pancreáticas , Neoplasias Pancreáticas , Animais , Camundongos , Diabetes Mellitus Experimental/metabolismo , Exocitose/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Ilhotas Pancreáticas/metabolismo , Neoplasias Pancreáticas/metabolismo
4.
J Tradit Chin Med ; 39(2): 191-198, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-32186041

RESUMO

OBJECTIVE: To determine the therapeutic effect and potential mechanism of Huatan Tongluo decoction on rats with collagen-induced arthritis. METHODS: Forty specific pathogen-free Wistar rats were selected, and 10 were randomly selected as the control (group 1). The remaining rats were injected intradermally with emulsified type II bovine collagen at the tail base and back, followed by a booster 7 d post first immunization. After establishing collagen-induced arthritis (CIA), rats were randomly divided into three groups (n = 10). The rats were treated orally for 30 d as follows: group 1, saline; group 2, model (saline); group 3, tripterygium polyglycoside (TP; 7.81 mg/kg, positive control); group 4, Huatan Tongluo decoction (HTTL; 7.5 g/kg). Body weight, ankle swelling and arthritis index were measured over the course of the study. The rats were sacrificed 30 d after treatment. Morphological changes in the synovium were observed by hematoxylin and eosin staining. Pannus formation and synovial thickness in the left ankle were observed by color Doppler ultrasoundVascular endothelial growth factor (VEGF) and VEGFR2 protein levels were measured by immunohistochemistry. VEGF/VEGFR2 mRNA levels were measured by real-time quantitative polymerase chain reaction. RESULTS: Compared with the model group, a significantly lower arthritis index was observed in the positive control group (P < 0.05) and HTTL group (P < 0.01), after treatment. Both positive control and HTTL reduced intra-articular pannus formation and synovial thickening. Furthermore, VEGF mRNA, and VEGFR2 protein and mRNA levels were significantly downregulated (P < 0.05) in the treatment groups. CONCLUSION: Inhibition of the expression of VEGF and VEGFR2 in synovial tissues and the formation of pannus and synovial hyperplasia may be part of the mechanism of HTTL for relieving the symptoms of rheumatoid arthritis in CIA rats.


Assuntos
Artrite Experimental/metabolismo , Artrite Experimental/patologia , Medicamentos de Ervas Chinesas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/patologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígenos CD34/metabolismo , Artrite Experimental/genética , Medicamentos de Ervas Chinesas/uso terapêutico , Masculino , RNA Mensageiro/genética , Ratos , Ratos Wistar , Membrana Sinovial/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
5.
Acta Pharmacol Sin ; 36(10): 1212-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26073330

RESUMO

Alternative splicing is a key process of multi-exonic gene expression during pre-mRNA maturation. In this process, particular exons of a gene will be included within or excluded from the final matured mRNA, and the resulting transcripts generate diverse protein isoforms. Recent evidence demonstrates that approximately 95% of human genes with multiple exons undergo alternative splicing during pre-mRNA maturation. Thus, alternative splicing plays a critical role in physiological processes and cell development programs, and.dysregulation of alternative splicing is highly associated with human diseases, such as cancer, diabetes and neurodegenerative diseases. In this review, we discuss the regulation of alternative splicing, examine the relationship between alternative splicing and human diseases, and describe several approaches that modify alternative splicing, which could aid in human disease diagnosis and therapy.


Assuntos
Processamento Alternativo , Diabetes Mellitus/genética , Descoberta de Drogas , Neoplasias/genética , Doenças Neurodegenerativas/genética , RNA Mensageiro/genética , Processamento Alternativo/efeitos dos fármacos , Animais , Diabetes Mellitus/tratamento farmacológico , Descoberta de Drogas/métodos , Marcadores Genéticos/genética , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico
6.
J Cell Biochem ; 115(2): 261-70, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24038283

RESUMO

Mitogen-activated kinase activating death domain containing protein (MADD) is abundantly expressed in cancer cells and necessary for maintaining cancer cell survival. However, this survival function of MADD is dependent upon its phosphorylation by protein kinase B (Akt). The tumour suppressor PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a lipid phosphatase that negatively regulates the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. The downstream targets of PTEN in triggering apoptosis have not yet been completely identified. Here, we report that MADD can act as a pro-apoptotic factor to initiate TRAIL-induced apoptosis when its phosphorylation is attenuated by PTEN. Our data show that tumor necrosis factor α-related apoptosis-inducing ligand (TRAIL) induced a reduction in MADD phosphorylation with a concomitant up-regulation of PTEN. Knock down of PTEN using a specific siRNA prevented TRAIL-induced reduction in pMADD levels. Surprisingly, Akt non-phosphorylated MADD translocated from the plasma membrane to cytoplasm where it bound to 14-3-3 and displaced 14-3-3 associated Bax, which translocated to mitochondria resulting in cytochrome c release. Taken together, our data reveal that PTEN can convey the death signal by preventing MADD phosphorylation by Akt.


Assuntos
Apoptose/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas 14-3-3/metabolismo , Linhagem Celular Tumoral , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Mitocôndrias/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Fator de Necrose Tumoral alfa/genética
7.
PLoS One ; 8(2): e56817, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23457619

RESUMO

The Map kinase Activating Death Domain containing protein (MADD) isoform of the IG20 gene is over-expressed in different types of cancer tissues and cell lines and it functions as a negative regulator of apoptosis. Therefore, we speculated that MADD might be over-expressed in human breast cancer tissues and that MADD knock-down might synergize with chemotherapeutic or TRAIL-induced apoptosis of breast cancer cells. Analyses of breast tissue microarrays revealed over-expression of MADD in ductal and invasive carcinomas relative to benign tissues. MADD knockdown resulted in enhanced spontaneous apoptosis in human breast cancer cell lines. Moreover, MADD knockdown followed by treatment with TRAIL or doxorubicin resulted in increased cell death compared to either treatment alone. Enhanced cell death was found to be secondary to increased caspase-8 activation. These data indicate that strategies to decrease MADD expression or function in breast cancer may be utilized to increase tumor cell sensitivity to TRAIL and doxorubicin induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/deficiência , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Doxorrubicina/farmacologia , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Sequência de Aminoácidos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Receptores de Morte Celular/metabolismo
8.
Thyroid ; 23(1): 70-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22998497

RESUMO

BACKGROUND: The IG20/MADD gene is overexpressed in thyroid cancer tissues and cell lines, and can contribute to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance. The ability of the MADD protein to resist TRAIL-induced apoptosis is dependent upon its phosphorylation by Akt. Interestingly, while TRAIL induces a significant reduction in the levels of phospho-Akt (pAkt) and phospho-MADD (pMADD) in TRAIL-sensitive cells, it fails to do so in TRAIL-resistant cells. In this study, we investigated if MADD phosphorylation by Akt was contributing to TRAIL resistance in thyroid cancer cells. METHODS: We determined the susceptibility of different thyroid cancer cell lines to TRAIL-induced apoptosis by fluorescence-activated cell sorting (FACS) analysis. We tested for various TRAIL resistance factors by FACS analyses or for IG20/MADD expression by quantitative reverse transcription-polymerase chain reaction. We determined the levels of pAkt and pMADD upon TRAIL treatment in thyroid cancer cells by Western blotting. We tested if down-modulation of IG20/MADD gene expression using shRNA or phosphorylation using a dominant negative Akt (DN-Akt) or pretreatment with LY294002, a PI3 kinase inhibitor, could help overcome TRAIL resistance. RESULT: BCPAP and TPC1 cells were susceptible, while KTC1 and FTC133 cells were resistant, to TRAIL-induced apoptosis. The differential susceptibility to TRAIL was not related to the levels of expression of death receptors, decoy receptors, or TRAIL. KTC1 and FTC133 cells showed higher levels of IG20/MADD expression relative to BCPAP and TPC1, and were rendered susceptible to TRAIL treatment upon IG20/MADD knockdown. Interestingly, upon TRAIL treatment, the pAkt and pMADD levels were reduced in TRAIL-sensitive BCPAP and TPC1 cells, while they remained unchanged in the resistant KTC1 and FTC133 cells. While expression of a constitutively active Akt in BCPAP and TPC1 cells rendered them resistant to TRAIL, pretreating KTC1 and FTC133 cells with LY294002 rendered them TRAIL-sensitive. Moreover, expression of a DN-Akt in KTC1 and FTC133 cells reduced the levels of pAkt and pMADD and sensitized them to TRAIL-induced apoptosis. CONCLUSION: Our results show that pMADD is an important TRAIL resistance factor in certain thyroid cancer cells and suggest that down-modulation of either IG20/MADD expression or phosphorylation can render TRAIL-resistant thyroid cancer cells sensitive to TRAIL.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cromonas/farmacologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores de Morte Celular/genética , Proteínas Recombinantes/farmacologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Receptores Chamariz do Fator de Necrose Tumoral/genética
9.
Am J Obstet Gynecol ; 205(4): 362.e12-25, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21855847

RESUMO

OBJECTIVE: The clinical utility of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in the treatment of established human malignancies is limited by the development of resistance to TRAIL. We hypothesized that knockdown of map-kinase activating death domain containing protein (MADD), a TRAIL-resistance factor, may overcome TRAIL resistance in ovarian cancer cells. STUDY DESIGN: MADD expression in resected ovarian cancer specimens and cell lines was quantified with the use of polymerase chain reaction. Sensitivity of ovarian cancer cell lines to TRAIL, with or without MADD knockdown, was assessed. RESULTS: MADD is expressed at relatively higher levels in human malignant ovarian cancer tissues and cell lines, compared with normal ovarian tissues. The cell lines OVCA429 and OVCAR3 were susceptible, and cell lines CAOV-3 and SKOV-3 were resistant to TRAIL. MADD knockdown in CAOV-3 cells, but not in SKOV-3 cells, conferred TRAIL sensitivity. Knockdown of cellular Fas-associated death domain-like interleukin-1 beta-converting enzyme-inhibitory protein (c-FLIP) in SKOV-3 cells increased spontaneous and TRAIL-induced apoptosis, which was further increased on MADD knockdown. CONCLUSION: MADD/c-FLIP(L) knockdown can render TRAIL-resistant ovarian cancer cells susceptible to TRAIL.


Assuntos
Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Neoplasias Ovarianas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Feminino , Humanos , Células Tumorais Cultivadas
10.
Ann N Y Acad Sci ; 1132: 276-82, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18567878

RESUMO

Current treatments for myasthenia gravis (MG) rely upon the administration of immunosuppressive agents which result in global, nonspecific attenuation of the immune response. An alternative approach would be to attempt to design therapies that specifically dampen autoreactivity without affecting general immunity. Recently, dendritic cells (DCs) have been shown to possess potent capabilities to tolerize T cells in an antigen-specific manner. We have observed that the selective activation of particular subsets of DCs utilizing granulocyte-macrophage colony-stimulating factor (GM-CSF) had profound effects on the induction of experimental autoimmune myasthenia gravis (EAMG). Specifically, treatment with GM-CSF effectively suppressed the induction of EAMG and down-modulated anti-AChR T cell and pathogenic antibody responses. These effects were associated with the activation of tolerogenic DCs, the enhanced production of suppressive cytokines, such as IL-10, and the mobilization of CD4(+)CD25(+) and FoxP3(+) regulatory T cells (Tregs). We have further shown that GM-CSF effectively ameliorates clinical disease severity in mice with active, ongoing EAMG. Based on these observations, we hypothesize that the selective activation of particular DC subsets in vivo using pharmacologic agents, like GM-CSF, can suppress ongoing anti-AChR immune responses by mobilizing antigen-specific Tregs capable of suppressing autoimmune MG.


Assuntos
Autoimunidade/imunologia , Miastenia Gravis Autoimune Experimental/imunologia , Animais , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Tolerância Imunológica/imunologia
11.
J Immunol ; 177(8): 5296-306, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015715

RESUMO

Dendritic cells (DCs) have the potential to activate or tolerize T cells in an Ag-specific manner. Although the precise mechanism that determines whether DCs exhibit tolerogenic or immunogenic functions has not been precisely elucidated, growing evidence suggests that DC function is largely dependent on differentiation status, which can be manipulated using various growth factors. In this study, we investigated the effects of mobilization of specific DC subsets-using GM-CSF and fms-like tyrosine kinase receptor 3-ligand (Flt3-L)-on the susceptibility to induction of experimental autoimmune myasthenia gravis (EAMG). We administered GM-CSF or Flt3-L to C57BL/6 mice before immunization with acetylcholine receptor (AChR) and observed the effect on the frequency and severity of EAMG development. Compared with AChR-immunized controls, mice treated with Flt3-L before immunization developed EAMG at an accelerated pace initially, but disease frequency and severity was comparable at the end of the observation period. In contrast, GM-CSF administered before immunization exerted a sustained suppressive effect against the induction of EAMG. This suppression was associated with lowered serum autoantibody levels, reduced T cell proliferative responses to AChR, and an expansion in the population of FoxP3+ regulatory T cells. These results highlight the potential of manipulating DCs to expand regulatory T cells for the control of autoimmune diseases such as MG.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fatores de Transcrição Forkhead , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Miastenia Gravis Autoimune Experimental/prevenção & controle , Linfócitos T Reguladores/citologia , Animais , Doenças Autoimunes/terapia , Comunicação Celular/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Imunização , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/terapia , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/imunologia
12.
Acta Pharmacol Sin ; 23(2): 138-42, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11866874

RESUMO

AIM: To examine the effect of sanggenon C on human polymorphonuclear leukocyte (PMN) adhesion to human synovial cell(HSC), and explore its mechanism. METHODS: Adhesion of PMN to HSC was measured by MTT colorimetry. Cell-ELISA and RT-PCR methods were used to examine the expression of adhesion molecules ICAM-1 and VCAM-1. Activation of nuclear factor-kappa B(NF-kappaB) was measured by electrophoretic mobility shift assays(EMSA) method. RESULTS: Sanggenon C effectively inhibited TNF-alpha (50 kU/L for 12 h) and IL-1beta (50 kU/L for 12 h) induced adhesion of PMN to HSC (IC50 27.29 nmol/L and 54.45 nmol/L, respectively) in a concentration-dependent manner. Adhesion molecule VCAM-1 surface protein and mRNA expression induced by TNF-alpha 50 kU/L were significantly inhibited by sanggenon C, nevertheless, for ICAM-1 only surface protein expression being inhibited. The activation of NF-kappaB was also extensively inhibited by sanggenon C. CONCLUSION: Sanggenon C inhibited TNF-alpha -stimulated PMN-HSC adhesion and expression of VCAM-1 by suppressing the activation of NF-kappaB.


Assuntos
Benzofuranos/farmacologia , Cromonas/farmacologia , Morus/química , Neutrófilos/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Separação Celular , Células Cultivadas , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , NF-kappa B/metabolismo , Neutrófilos/citologia , Raízes de Plantas/química , Membrana Sinovial/citologia , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
13.
Yao Xue Xue Bao ; 37(2): 103-7, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12579952

RESUMO

AIM: To investigate the effect of meloxicam on human polymorphonuclear leukocyte (PMN) adhesion to human synovial cell (HSC), and to explore its mechanism. METHODS: MTT colorimetry was used to determine the adhesion effect of PMN to HSC. Cell-ELISA and RT-PCR methods were used to determine the expression of ICAM-1 and VCAM-1. Nuclear transcription factor-kappa B (NF-kappa B) was measured by electrophoretic mobility shift assay (EMSA) method. RESULTS: Meloxicam was found to effectively inhibit TNF-alpha (50 u.mL-1 for 12 h) and IL-1 beta (50 u.mL-1 for 12 h)-induced adhesion of PMN to HSC (IC50 3.38 x 10(-7) mol.L-1 and 3.56 x 10(-6) mol.L-1, respectively) in a concentration-dependent manner. ICAM-1 protein and mRNA expression induced by TNF-alpha (50 u.mL-1) were inhibited by meloxicam at 1 x 10(-6)-1 x 10(-5) mol.L-1. The activation of NF-kappa B was also inhibited by meloxicam at 1 x 10(-6)-1 x 10(-5) mol.L-1. CONCLUSION: These results suggest that meloxicam inhibit TNF-alpha stimulated PMN-HSC adhesion and expression of ICAM-1 by suppressing the activity of NF-kappa B.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Adesão Celular/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Membrana Sinovial/citologia , Tiazinas/farmacologia , Tiazóis/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Meloxicam , NF-kappa B/metabolismo , Neutrófilos/fisiologia , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA