Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
The Korean Journal of Physiology and Pharmacology ; : 1-15, 2018.
Artigo em Inglês | WPRIM | ID: wpr-727943

RESUMO

Inflammasomes are intracellular multiprotein complexes that coordinate anti-pathogenic host defense during inflammatory responses in myeloid cells, especially macrophages. Inflammasome activation leads to activation of caspase-1, resulting in the induction of pyroptosis and the secretion of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18. Although the inflammatory response is an innate host defense mechanism, chronic inflammation is the main cause of rheumatic diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), ankylosing spondylitis (AS), and Sjögren's syndrome (SS). Since rheumatic diseases are inflammatory/autoimmune disorders, it is reasonable to hypothesize that inflammasomes activated during the inflammatory response play a pivotal role in development and progression of these diseases. Indeed, previous studies have provided important observations that inflammasomes are actively involved in the pathogenesis of inflammatory/autoimmune rheumatic diseases. In this review, we summarize the current knowledge on several types of inflammasomes during macrophage-mediated inflammatory responses and discuss recent research regarding the role of inflammasomes in the pathogenesis of inflammatory/autoimmune rheumatic diseases. This avenue of research could provide new insights for the development of promising therapeutics to treat inflammatory/autoimmune rheumatic diseases.


Assuntos
Artrite Reumatoide , Autoimunidade , Citocinas , Inflamassomos , Inflamação , Interleucina-18 , Interleucinas , Lúpus Eritematoso Sistêmico , Macrófagos , Complexos Multiproteicos , Células Mieloides , Piroptose , Doenças Reumáticas , Espondilite Anquilosante
2.
The Korean Journal of Parasitology ; : 711-717, 2016.
Artigo em Inglês | WPRIM | ID: wpr-72763

RESUMO

Toxoplasma gondii is an obligate intracellular parasite that stimulates production of high levels of proinflammatory cytokines, which are important for innate immunity. NLRs, i.e., nucleotide-binding oligomerization domain (NOD)-like receptors, play a crucial role as innate immune sensors and form multiprotein complexes called inflammasomes, which mediate caspase-1-dependent processing of pro-IL-1β. To elucidate the role of inflammasome components in T. gondii-infected THP-1 macrophages, we examined inflammasome-related gene expression and mechanisms of inflammasome-regulated cytokine IL-1β secretion. The results revealed a significant upregulation of IL-1β after T. gondii infection. T. gondii infection also upregulated the expression of inflammasome sensors, including NLRP1, NLRP3, NLRC4, NLRP6, NLRP8, NLRP13, AIM2, and NAIP, in a time-dependent manner. The infection also upregulated inflammasome adaptor protein ASC and caspase-1 mRNA levels. From this study, we newly found that T. gondii infection regulates NLRC4, NLRP6, NLRP8, NLRP13, AIM2, and neuronal apoptosis inhibitor protein (NAIP) gene expressions in THP-1 macrophages and that the role of the inflammasome-related genes may be critical for mediating the innate immune responses to T. gondii infection.


Assuntos
Apoptose , Citocinas , Expressão Gênica , Imunidade Inata , Inflamassomos , Macrófagos , Complexos Multiproteicos , Negociação , Neurônios , Parasitos , RNA Mensageiro , Toxoplasma , Regulação para Cima
3.
Protein & Cell ; (12): 749-760, 2016.
Artigo em Inglês | WPRIM | ID: wpr-757388

RESUMO

The BCCIP (BRCA2- and CDKN1A-interacting protein) is an important cofactor for BRCA2 in tumor suppression. Although the low expression of BCCIP is observed in multiple clinically diagnosed primary tumor tissues such as ovarian cancer, renal cell carcinoma and colorectal carcinoma, the mechanism of how BCCIP is regulated in cells is still unclear. The human INO80/YY1 chromatin remodeling complex composed of 15 subunits catalyzes ATP-dependent sliding of nucleosomes along DNA. Here, we first report that BCCIP is a novel target gene of the INO80/YY1 complex by presenting a series of experimental evidence. Gene expression studies combined with siRNA knockdown data locked candidate genes including BCCIP of the INO80/YY1 complex. Silencing or over-expressing the subunits of the INO80/YY1 complex regulates the expression level of BCCIP both in mRNA and proteins in cells. Also, the functions of INO80/YY1 complex in regulating the transactivation of BCCIP were confirmed by luciferase reporter assays. Chromatin immunoprecipitation (ChIP) experiments clarify the enrichment of INO80 and YY1 at +0.17 kb downstream of the BCCIP transcriptional start site. However, this enrichment is significantly inhibited by either knocking down INO80 or YY1, suggesting the existence of both INO80 and YY1 is required for recruiting the INO80/YY1 complex to BCCIP promoter region. Our findings strongly indicate that BCCIP is a potential target gene of the INO80/YY1 complex.


Assuntos
Humanos , Proteínas de Ligação ao Cálcio , Genética , Metabolismo , Proteínas de Ciclo Celular , Genética , Metabolismo , Montagem e Desmontagem da Cromatina , Fisiologia , DNA Helicases , Genética , Metabolismo , Células HeLa , Complexos Multiproteicos , Genética , Metabolismo , Proteínas Nucleares , Genética , Metabolismo , Regiões Promotoras Genéticas , Fisiologia , Transcrição Gênica , Fisiologia , Fator de Transcrição YY1 , Genética , Metabolismo
4.
Protein & Cell ; (12): 878-887, 2016.
Artigo em Inglês | WPRIM | ID: wpr-757348

RESUMO

Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) integrates signals from growth factors, cellular energy levels, stress and amino acids to control cell growth and proliferation through regulating translation, autophagy and metabolism. Here we determined the cryo-electron microscopy structure of human mTORC1 at 4.4 Å resolution. The mTORC1 comprises a dimer of heterotrimer (mTOR-Raptor-mLST8) mediated by the mTOR protein. The complex adopts a hollow rhomboid shape with 2-fold symmetry. Notably, mTORC1 shows intrinsic conformational dynamics. Within the complex, the conserved N-terminal caspase-like domain of Raptor faces toward the catalytic cavity of the kinase domain of mTOR. Raptor shows no caspase activity and therefore may bind to TOS motif for substrate recognition. Structural analysis indicates that FKBP12-Rapamycin may generate steric hindrance for substrate entry to the catalytic cavity of mTORC1. The structure provides a basis to understand the assembly of mTORC1 and a framework to characterize the regulatory mechanism of mTORC1 pathway.


Assuntos
Humanos , Linhagem Celular , Microscopia Crioeletrônica , Métodos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Química , Estrutura Quaternária de Proteína , Serina-Treonina Quinases TOR , Química
5.
Journal of Experimental Hematology ; (6): 1086-1090, 2016.
Artigo em Chinês | WPRIM | ID: wpr-246812

RESUMO

<p><b>UNLABELLED</b>Objective:To explore the influence of co-inhibiting mTORC2 and HSP90 on the proliferation and apoptosis of multiple myeloma(MM) cell line U266.</p><p><b>METHODS</b>During culture, the human MM cell line U266 were treated with 20 nmol/L of rapamycin, 600 nmol/L 17-AAG, 20 nmol/L of rapamycin + 600 nmol/L 17-AGG and phosphate-buffered saline (PBS), then the growth inhibition rate, morphologic changes, apoptosis rate and the expression of caspase 3 and ATK protein in U266 cells were compared and analyzed.</p><p><b>RESULTS</b>The rapamycin and 17-AAG both could inhibit the growth of U266 cells, while the inhibitory effect of rapamycin in combination with 17-AAG on growth of U266 cells was significantly higher them that of rapamycin and 17-AAG alone and control (PBS); the apoptosis rate of U266 cells treated with rapamycin, 17-AAG and their combination was higher than that of control PBS groups, and the efficacy of 2 drug conbination was higher than that of control PBS group, and the efficacy of 2 drug combination was superior to single drug. The expression levels of caspase 3 and ATK in U266 cells treated with rapamycin, 17-AAG and their combination were higher and lower than those in control group respectively, and the efficacy of 2 drug combination was superior to signle drug. There were significant difference between them (P<0.05).</p><p><b>CONCLUSION</b>The co-inhibition of mTORC2 and HSP90 can suppress the proliferation and induce the apoptosis of MM cells.</p>


Assuntos
Humanos , Apoptose , Benzoquinonas , Caspase 3 , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Choque Térmico HSP90 , Lactamas Macrocíclicas , Alvo Mecanístico do Complexo 2 de Rapamicina , Mieloma Múltiplo , Complexos Multiproteicos , Sirolimo , Serina-Treonina Quinases TOR
6.
Rev. bras. anestesiol ; 65(3): 222-229, May-Jun/2015. tab
Artigo em Inglês | LILACS | ID: lil-748917

RESUMO

BACKGROUND AND OBJECTIVES: In this study, the aim was to compare postoperative analgesia effects of the administration of ultrasound-guided interscalene brachial plexus block and intra-articular bupivacaine carried out with bupivacaine. METHODS: In the first group of patients 20 mL 0.25% bupivacaine and ultrasound-guided interscalene brachial plexus block (ISPB) were applied, while 20 mL 0.25% bupivacaine was given via intra-articular (IA) administration to the second group patients after surgery. Patients in the third group were considered the control group and no block was performed. Patient-controlled analgesia (PCA) with morphine was used in all three groups for postoperative analgesia. RESULTS: In the ISPB group, morphine consumption in the periods between 0-4, 6-12 and 12-24 postoperative hours and total consumption within 24 h was lower than in the other two groups. Morphine consumption in the IA group was lower than in the control group in the period from 0 to 6 h and the same was true for total morphine consumption in 24 h. Postoperative VASr scores in the ISPB group were lower than both of the other groups in the first 2 h and lower than the control group in the 4th and 6th hours (p < 0.05). In the IA group, VASr and VASm scores in the 2nd, 4th and 6th hours were lower than in the control group (p < 0.05). CONCLUSION: Interscalene brachial plexus block was found to be more effective than intra-articular local anesthetic injection for postoperative analgesia. .


JUSTIFICATIVA E OBJETIVOS: Comparar os efeitos na analgesia no pós-operatório da administração de bloqueio do plexo braquial por via interescalênica guiado por ultrassom e bupivacaína intra-articular, feito com bupivacaína. MÉTODOS: No primeiro grupo de pacientes, 20 mL de bupivacaína a 0,25% e bloqueio do plexo braquial por via interescalênica guiado por ultrassom (BPBI) foram administrados, enquanto 20 mL de bupivacaína a 0,25% foram administrados por via intra-articular (IA) ao segundo grupo de pacientes após a cirurgia. Os pacientes do terceiro grupo foram considerados grupo controle e nenhum bloqueio foi feito. Analgesia controlada pelo paciente (ACP) com morfina foi usada nos três grupos para analgesia pós-operatória. RESULTADOS: No grupo BPBI, o consumo de morfina nos períodos entre 0-4, 6-12 e 12-24 horas após a cirurgia e o consumo total em 24 horas foram mais baixos do que nos outros dois grupos. O consumo de morfina no grupo IA foi menor do que no grupo controle no período de 0-6 horas, como também foi menor o consumo total de morfina em 24 horas. Os escores EVAr no pós-operatório do grupo BPBI foram menores do que os escores dos dois outros grupos nas primeiras duas horas e menores do que os do grupo controle nos períodos de 4 e 6 horas (p < 0,05). No grupo IA, os escores EVAr e EVAm nos períodos de 2, 4 e 6 horas foram menores do que no grupo controle (p < 0,05). CONCLUSÃO: O bloqueio do plexo braquial por via interescalênica mostrou ser mais eficaz do que a injeção intra-articular de anestésico local para analgesia pós-operatória. .


JUSTIFICACIÓN Y OBJETIVOS: En este estudio, nuestro objetivo fue comparar en el período postoperatorio los efectos analgésicos de la administración de la bupivacaína en el bloqueo del plexo braquial por vía interescalénica guiado por ecografía y bupivacaína intraarticular. MÉTODOS: En el primer grupo de pacientes se administraron 20 mL de bupivacaína al 0,25% y se llevó a cabo el bloqueo del plexo braquial por vía interescalénica (BPBI) guiado por ecografía, mientras que al segundo grupo de pacientes se le administraron 20 mL de bupivacaína al 0,25% por vía intraarticular (IA) tras la cirugía. Los pacientes del tercer grupo fueron considerados como grupo control y en ellos no se realizó ningún bloqueo. La analgesia controlada por el paciente con morfina se usó en los 3 grupos para la analgesia postoperatoria. RESULTADOS: En el grupo BPBI, el consumo de morfina en los períodos entre 0-4, 6-12 y 12-24 h del postoperatorio y el consumo total en 24 h fueron más bajos que en los otros 2 grupos. El consumo de morfina en el grupo IA fue menor que en el grupo control en el período de 0-6 h, como también fue menor el consumo total de morfina en 24 h. Las puntuaciones EVAr en el postoperatorio del grupo BPBI fueron menores que las de los otros 2 grupos en las primeras 2 h y menores que los del grupo control en los períodos de 4 y 6 h (p < 0,05). En el grupo IA, las puntuaciones EVAr y EVAm en los períodos de 2, 4 y 6 h fueron menores que en el grupo control (p < 0,05). CONCLUSIÓN: El BPBI mostró ser más eficaz que la inyección intraarticular de anestésico local para analgesia postoperatoria. .


Assuntos
Dineínas/metabolismo , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Motores Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Dineínas/química , Dineínas/isolamento & purificação , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico
7.
Experimental & Molecular Medicine ; : e155-2015.
Artigo em Coreano | WPRIM | ID: wpr-147137

RESUMO

The long-term storage of memory requires the finely tuned coordination of intracellular signaling with the transcriptional, translational and epigenetic regulations of gene expression. Among the epigenetic mechanisms, however, we know relatively little about the involvement of chromatin remodeling-dependent control of gene expression in cognitive brain functions, compared with our knowledge of other such mechanisms (for example, histone modifications and DNA methylation). A few recent studies have implicated the Brm/Brg-associated factor (BAF) chromatin-remodeling complex, a mammalian homolog of the yeast Swi/Snf complex, in neuronal structural/functional plasticity and memory formation. The BAF complex was previously known to have a critical role in neurodevelopment, but these recent findings indicate that it also contributes to both cognitive functions in the adult brain and human mental disorders characterized by intellectual disability. In this review, we provide a brief overview of the BAF complexes, introduce recent research findings that link their functions to memory formation, and speculate on the yet-unknown molecular mechanisms that may be relevant to these processes.


Assuntos
Animais , Humanos , Actinas/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Aprendizagem , Memória , Complexos Multiproteicos/metabolismo , Neurônios/metabolismo , Ligação Proteica , Transdução de Sinais , Fatores de Transcrição/metabolismo
8.
Journal of Experimental Hematology ; (6): 1277-1281, 2015.
Artigo em Chinês | WPRIM | ID: wpr-274051

RESUMO

<p><b>OBJECTIVE</b>To investigate the expression of CSN complex (COP9 signal some subunits) in the patients with acute promyelocytic leukemia (APL) and its significance in the ATRA-induced APL differentiation.</p><p><b>METHODS</b>Using the NB4 cells as a model, morphologic observation and myeloid differentiation marker CD11b detection were used to monitor ATRA-induced APL differentiation, the expression of CSN complex in cell differentiation was detected by Western blot and reverse transcription real time fluorescent quantitative PCR (RT-qPCR) method. RT-qPCR was also used to detect the relative expression level of COP9 signalosome subunits in the APL patients and remission after treatment.</p><p><b>RESULTS</b>ATRA could obviously enhance CD11b expression; the cell morphology showed obvious differentiation characteristics. During the differentiation, the expression of COP9 signalosome subunits was down-regulated by ATRA. Meanwhile, the CSN expression level in newly diagnosed APL patients was much higher than that in controls (non-leukemia) (P < 0.05). The level of CSN expression was obviously down-regulated when APL patients achieved complete remission.</p><p><b>CONCLUSION</b>The high CSN expression level in APL patients can be down-regulated by ATRA. CSN complex may have a significant effect on the pathogenesis and therapy of APL.</p>


Assuntos
Humanos , Complexo do Signalossomo COP9 , Diferenciação Celular , Linhagem Celular Tumoral , Regulação para Baixo , Leucemia Promielocítica Aguda , Metabolismo , Complexos Multiproteicos , Metabolismo , Peptídeo Hidrolases , Metabolismo , Tretinoína , Farmacologia
9.
Chinese Journal of Hematology ; (12): 780-784, 2015.
Artigo em Chinês | WPRIM | ID: wpr-296153

RESUMO

<p><b>OBJECTIVE</b>To explore apoptosis of multiple myeloma (MM) cells and its mechanism by the combined inhibition of mTORC2 signaling pathway and heat shock protein 90.</p><p><b>METHODS</b>The effects of Rapamycin, 17-AAG and the combination on proliferation of MM cell lines U266 and KM3 were assessed using MTT at different time points (0, 8, 24, 48 hour). Cell apoptosis and cell cycle distribution were measured by flow cytometry. The specific proteins p-AKT (ser473), p-AKT (thr450), p-S6 (S235/236) and AKT were detected by Western blotting.</p><p><b>RESULTS</b>Rapamycin, 17- AAG and the combination suppressed the proliferation of MM cell lines U266 and KM3, especially the combination of Rapamycin and 17-AAG synergistically inhibited the proliferation (P<0.05); Rapamycin induced G1 arrest both at 24 and 48 hours, 17-AAG also induced G1 arrest, especially at 48 hours (P<0.01); Rapamycin, 17-AAG alone decreased the expression of AKT and induced MM cell apoptosis to some extent (P<0.01); Chronic rapamycin treatment inhibited mTORC2; Inhibition of both mTORC2 and chaper on pathways degraded AKT and induced MM cell apoptosis, which was significantly higher than that of any single agent (P<0.01).</p><p><b>CONCLUSION</b>Inhibition of both mTORC2 and chaper on pathways decreased the expression of AKT to induce apoptosis of MM cells in vitro.</p>


Assuntos
Humanos , Apoptose , Benzoquinonas , Farmacologia , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP90 , Metabolismo , Lactamas Macrocíclicas , Farmacologia , Alvo Mecanístico do Complexo 2 de Rapamicina , Mieloma Múltiplo , Patologia , Complexos Multiproteicos , Metabolismo , Proteínas Proto-Oncogênicas c-akt , Metabolismo , Transdução de Sinais , Sirolimo , Farmacologia , Serina-Treonina Quinases TOR , Metabolismo
10.
Experimental & Molecular Medicine ; : e143-2015.
Artigo em Inglês | WPRIM | ID: wpr-42472

RESUMO

An F-box protein, beta-TrCP recognizes substrate proteins and destabilizes them through ubiquitin-dependent proteolysis. It regulates the stability of diverse proteins and functions as either a tumor suppressor or an oncogene. Although the regulation by beta-TrCP has been widely studied, the regulation of beta-TrCP itself is not well understood yet. In this study, we found that the level of beta-TrCP1 is downregulated by various protein kinase inhibitors in triple-negative breast cancer (TNBC) cells. A PI3K/mTOR inhibitor PI-103 reduced the level of beta-TrCP1 in a wide range of TNBC cells in a proteasome-dependent manner. Concomitantly, the levels of c-Myc and cyclin E were also downregulated by PI-103. PI-103 reduced the phosphorylation of beta-TrCP1 prior to its degradation. In addition, knockdown of beta-TrCP1 inhibited the proliferation of TNBC cells. We further identified that pharmacological inhibition of mTORC2 was sufficient to reduce the beta-TrCP1 and c-Myc levels. These results suggest that mTORC2 regulates the stability of beta-TrCP1 in TNBC cells and targeting beta-TrCP1 is a potential approach to treat human TNBC.


Assuntos
Feminino , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Ciclina E/genética , Relação Dose-Resposta a Droga , Furanos/farmacologia , Técnicas de Silenciamento de Genes , Modelos Biológicos , Complexos Multiproteicos/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Piridinas/farmacologia , Pirimidinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/genética , Proteínas Contendo Repetições de beta-Transducina/genética
11.
Indian J Biochem Biophys ; 2014 Oct; 51(5): 335-342
Artigo em Inglês | IMSEAR | ID: sea-154261

RESUMO

Perception of molecular mechanism would provide potent additional knowledge on mammalian membrane proteins involved in causing diseases. In human, syntaxin-3 (STX3) is a significant apical targeting protein in the epithelial membrane and in exocytosis process; it also acts as a vesicle transporter by cellular receptor in neutrophils, which is crucial for protein trafficking event. Structurally, syntaxin-3 has hydrophobic domain at carboxyl terminus that directs itself to intra-cellular compartments. In addition, the experimental structure of STX3 is not available and no mutational study has been carried out with natural variants of proteins. Moreover, there is no evidence so far for the natural variant Val286 of STX3 causing any diseases. Hence, in the present study, analyses of residue-based properties of the homology model STX3 were carried out along with mutations at carboxyl terminus of STX3 by implementing protein engineering and in silico approaches. The model structure of STX3 was constructed adopting Modeller v9.11 and the aggregation propensity was analyzed with BioLuminate tool. The results showed that there was reduction in aggregation propensity with point mutation at Val286, instead of Ile, resulting into increasing the structural stability of STX3. In conclusion, the Ccap exposed residue would be a suitable position for further mutational studies, particularly with Val286 of STX3 in human. This approach could gainfully be applied to STX3 for efficient drug designing which would be a valuable target in the cancer treatment.


Assuntos
Simulação por Computador , Humanos , Modelos Químicos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/ultraestrutura , Mutagênese Sítio-Dirigida , Mutação/genética , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/ultraestrutura , Homologia de Sequência de Aminoácidos
12.
Braz. j. med. biol. res ; 47(4): 307-3015, 8/4/2014. graf
Artigo em Inglês | LILACS | ID: lil-705765

RESUMO

Dye exclusion tests are used to determine the number of live and dead cells. These assays are based on the principle that intact plasma membranes in live cells exclude specific dyes, whereas dead cells do not. Although widely used, the trypan blue (TB) exclusion assay has limitations. The dye can be incorporated by live cells after a short exposure time, and personal reliability, related to the expertise of the analyst, can affect the results. We propose an alternative assay for evaluating cell viability that combines the TB exclusion test and the high sensitivity of the flow cytometry technique. Previous studies have demonstrated the ability of TB to emit fluorescence when complexed with proteins. According to our results, TB/bovine serum albumin and TB/cytoplasmic protein complexes emit fluorescence at 660 nm, which is detectable by flow cytometry using a 650-nm low-pass band filter. TB at 0.002% (w/v) was defined as the optimum concentration for distinguishing unstained living cells from fluorescent dead cells, and fluorescence emission was stable for 30 min after cell treatment. Although previous studies have shown that TB promotes green fluorescence quenching, TB at 0.002% did not interfere with green fluorescence in human live T-cells stained with anti-CD3/fluorescein isothiocyanate (FITC) monoclonal antibody. We observed a high correlation between the percentage of propidium iodide+CD3/FITC+ and TB+CD3/FITC+ cells, as well as similar double-stained cell profiles in flow cytometry dot-plot graphs. Taken together, the results indicate that a TB exclusion assay by flow cytometry can be employed as an alternative tool for quick and reliable cell viability analysis.


Assuntos
Humanos , Adulto Jovem , /sangue , Citometria de Fluxo/normas , Leucócitos Mononucleares/metabolismo , Azul Tripano , Contagem de Células , Separação Celular , Sobrevivência Celular , Membrana Celular/fisiologia , Fluorescência , Imunofenotipagem , Indicadores e Reagentes/normas , Complexos Multiproteicos/normas , Competência Profissional , Propídio/normas , Coloração e Rotulagem , Soroalbumina Bovina/normas
13.
Clinical and Experimental Vaccine Research ; : 5-11, 2014.
Artigo em Inglês | WPRIM | ID: wpr-36958

RESUMO

Inflammasomes are cytosolic multiprotein complexes that sense microbial motifs or cellular stress and stimulate caspase-1-dependent cytokine secretion and cell death. Recently, it has become increasingly evident that both DNA and RNA viruses activate inflammasomes, which control innate and adaptive immune responses against viral infections. In addition, recent studies suggest that certain microbiota induce inflammasomes-dependent adaptive immunity against influenza virus infections. Here, we review recent advances in research into the role of inflammasomes in antiviral immunity.


Assuntos
Imunidade Adaptativa , Morte Celular , Citosol , Células Dendríticas , DNA , Inflamassomos , Vacinas contra Influenza , Influenza Humana , Metagenoma , Microbiota , Complexos Multiproteicos , Orthomyxoviridae , Vírus de RNA
14.
Protein & Cell ; (12): 171-177, 2014.
Artigo em Inglês | WPRIM | ID: wpr-757517

RESUMO

Mammalian target of rapamycin (mTOR) plays essential roles in cell proliferation, survival and metabolism by forming at least two functional distinct multi-protein complexes, mTORC1 and mTORC2. External growth signals can be received and interpreted by mTORC2 and further transduced to mTORC1. On the other hand, mTORC1 can sense inner-cellular physiological cues such as amino acids and energy states and can indirectly suppress mTORC2 activity in part through phosphorylation of its upstream adaptors, IRS-1 or Grb10, under insulin or IGF-1 stimulation conditions. To date, upstream signaling pathways governing mTORC1 activation have been studied extensively, while the mechanisms modulating mTORC2 activity remain largely elusive. We recently reported that Sin1, an essential mTORC2 subunit, was phosphorylated by either Akt or S6K in a cellular context-dependent manner. More importantly, phosphorylation of Sin1 at T86 and T398 led to a dissociation of Sin1 from the functional mTORC2 holo-enzyme, resulting in reduced Akt activity and sensitizing cells to various apoptotic challenges. Notably, an ovarian cancer patient-derived Sin1-R81T mutation abolished Sin1-T86 phosphorylation by disrupting the canonical S6K-phoshorylation motif, thereby bypassing Sin1-phosphorylation-mediated suppression of mTORC2 and leading to sustained Akt signaling to promote tumorigenesis. Our work therefore provided physiological and pathological evidence to reveal the biological significance of Sin1 phosphorylation-mediated suppression of the mTOR/Akt oncogenic signaling, and further suggested that misregulation of this process might contribute to Akt hyper-activation that is frequently observed in human cancers.


Assuntos
Animais , Humanos , Proteínas Adaptadoras de Transdução de Sinal , Metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Modelos Biológicos , Complexos Multiproteicos , Metabolismo , Fosforilação , Fosfotreonina , Metabolismo , Serina-Treonina Quinases TOR , Metabolismo
15.
Journal of Experimental Hematology ; (6): 1476-1479, 2014.
Artigo em Chinês | WPRIM | ID: wpr-340475

RESUMO

The inflammasome is a group of multiprotein complexes in the cytoplasm, which can activate caspase-1 that mediates the maturation and release of IL-1β, IL-18, IL-33 and other pro-inflammatory cytokines.NALP1 (NACHT leucine-rich-repeat protein 1), also known as NLRP1, is the first one of the identified complex inflammasomes with definite ligands mainly involved in the activation of inflammasome assembly and the formation of apoptotic bodies. Moreover, it was also found that NLRP1 plays an important biological role in the development of acute leukemia, the bone marrow hematopoietic stem cell apoptosis and other blood diseases. This review briefly summarizes the structure, activation mechanism, regulation and the role of NLRP1 in the hematopoietic system.


Assuntos
Humanos , Proteínas Adaptadoras de Transdução de Sinal , Metabolismo , Apoptose , Proteínas Reguladoras de Apoptose , Metabolismo , Citocinas , Doenças Hematológicas , Metabolismo , Patologia , Inflamassomos , Complexos Multiproteicos
16.
Journal of Experimental Hematology ; (6): 1063-1068, 2013.
Artigo em Chinês | WPRIM | ID: wpr-283981

RESUMO

Mammalian target of rapamycin complex (mTORC) is an important center for regulating cellular growth, survival and metabolism. mTORC plays a vital role in maintenance of normal physiological activities and homeostasis in organism. According to protein components, mTORC can be divided into two distinct protein complexes: mTORC1 and mTORC2. The main protein components of mTORC2 include mTOR, Rictor, mLST8, Deptor, mSin1, Protor and Hsp70. By means of activating AKT, PKCα, SGK1 and so on, the mTORC regulates many vital activities:embryonic development, cytoskeletal reconstitution,cell migration and protein post-translational modification. The abnormality of mTORC2 signaling pathway has been confirmed to be associated with tumorigenesis, therefore, further understanding the components, functions and signalling pathway of mTORC2 will provide a new insights in developing targeted cancer therapy. In this review, the structure and signalling pathway of mTORC2 and its roles in hematological malignancies are discussed and summarised.


Assuntos
Animais , Humanos , Neoplasias Hematológicas , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos , Transdução de Sinais , Serina-Treonina Quinases TOR
17.
Chinese Journal of Cancer ; (12): 427-433, 2013.
Artigo em Inglês | WPRIM | ID: wpr-320585

RESUMO

Mammalian target of rapamycin (mTOR) is aberrantly activated in many cancer types, and two rapamycin derivatives are currently approved by the Food and Drug Administration (FDA) of the United States for treating renal cell carcinoma. Mechanistically, mTOR is hyperactivated in human cancers either due to the genetic activation of its upstream activating signaling pathways or the genetic inactivation of its negative regulators. The tumor suppressor liver kinase B1 (LKB1), also known as serine/threonine kinase 11 (STK11), is involved in cell polarity, cell detachment and adhesion, tumor metastasis, and energetic stress response. A key role of LKB1 is to negatively regulate the activity of mTOR complex 1 (mTORC1). This review summarizes the molecular basis of this negative interaction and recent research progress in this area.


Assuntos
Animais , Feminino , Humanos , Proteínas Quinases Ativadas por AMP , Metabolismo , Adenocarcinoma , Tratamento Farmacológico , Metabolismo , Antibióticos Antineoplásicos , Usos Terapêuticos , Modelos Animais de Doenças , Neoplasias do Endométrio , Tratamento Farmacológico , Metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Metabolismo , Fosfatidilinositol 3-Quinases , Metabolismo , Proteínas Serina-Treonina Quinases , Metabolismo , Proteínas Proto-Oncogênicas c-akt , Metabolismo , Transdução de Sinais , Sirolimo , Usos Terapêuticos , Serina-Treonina Quinases TOR , Metabolismo , Proteínas Supressoras de Tumor , Metabolismo
18.
National Journal of Andrology ; (12): 1068-1071, 2013.
Artigo em Chinês | WPRIM | ID: wpr-267987

RESUMO

<p><b>OBJECTIVE</b>To investigate the roles of the mammalian target of rapamycin-1 and -2 (mTORC1 and TORC2) in the proliferation and apoptosis of prostate cancer 22RV1 cells.</p><p><b>METHODS</b>After silencing mTORC1 and TORC2, we examined the proliferation and apoptosis of prostate cancer 22RV1 cells by methylthiazol tetrazolium (MTT) assay and flow cytometry, respectively, and detected the expressions of the androgen receptor (AR) and Akt phosphorylation in the prostate cancer 22RV1 cells by Western blot after transfecting Raptor-siRNA and Rictor-siRNA to the 22RV1 cells.</p><p><b>RESULTS</b>MTT showed that the prostate cancer 22RV1 cells had no significant change in the growth rate after mTORC1 silence (P > 0.05), but their proliferation was markedly inhibited after mTORC2 silence (P < 0.01). Flow cytometry revealed a dramatic increase in the apoptosis of the 22RV1 cells after mTORC1 silence (P < 0.01), but no obvious change after mTORC2 silence (P > 0.05). Western blot exhibited that mTORC1 silence significantly increased the expression of AR and Akt phosphorylation (P < 0.05), while mTORC2 silence markedly decreased them (P < 0.05).</p><p><b>CONCLUSION</b>mTORC2 is not only required for the survival of prostate cancer 22RV1 cells, but also a promising therapeutic target of prostate cancer.</p>


Assuntos
Humanos , Masculino , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos , Metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Metabolismo , Receptores Androgênicos , Metabolismo , Sirolimo , Farmacologia , Serina-Treonina Quinases TOR , Metabolismo
19.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 810-816, 2013.
Artigo em Inglês | WPRIM | ID: wpr-251389

RESUMO

Autophagy is a conserved and programmed catabolic process that degrades damaged proteins and organelles. But the underlying mechanism and functions of autophagy in the ischemia-reperfusion (IR)-induced injury are unknown. In this study, we employed simulated IR of N2a cells as an in vitro model of IR injury to the neurons and monitored autophagic processes. It was found that the levels of Beclin-1 (a key molecule of autophay complex, Beclin-1/class III PI3K) and LC-3II (an autophagy marker) were remarkably increased with time during the process of ischemia and the process of reperfusion after 90 min of ischemia, while the protein kinases p70S6K and mTOR which are involved in autophagy regulation showed delayed inactivation after reperfusion. Administration of 3-methyladenine (3MA), an inhibitor of class III PI3K, abolished autophagy during reperfusion, while employment of rapamycin, an inhibitor of mTORC1 (normally inducing autophagy), surprisingly weakened the induction of autophagy during reperfusion. Analyses of mitochondria function by relative cell viability demonstrated that autophagy inhibition by 3-MA attenuated the decline of mitochondria function during reperfusion. Our data demonstrated that there were two distinct dynamic patterns of autophagy during IR-induced N2a injury, Beclin-1/class III PI3K complex-dependent and mTORC1-dependent. Inhibition of over-autophagy improved cell survival. These suggest that targeting autophagy therapy will be a novel strategy to control IR-induced neuronal damage.


Assuntos
Animais , Camundongos , Adenina , Farmacologia , Proteínas Reguladoras de Apoptose , Genética , Metabolismo , Autofagia , Proteína Beclina-1 , Linhagem Celular Tumoral , Sobrevivência Celular , Alvo Mecanístico do Complexo 1 de Rapamicina , Mitocôndrias , Metabolismo , Complexos Multiproteicos , Metabolismo , Neurônios , Metabolismo , Fármacos Neuroprotetores , Farmacologia , Fosfatidilinositol 3-Quinases , Metabolismo , Traumatismo por Reperfusão , Metabolismo , Sirolimo , Farmacologia , Serina-Treonina Quinases TOR , Metabolismo
20.
Chinese Journal of Cancer ; (12): 8-18, 2012.
Artigo em Inglês | WPRIM | ID: wpr-294462

RESUMO

The mammalian target of rapamycin (mTOR), a serine/threonine protein kinase, acts as a "master switch" for cellular anabolic and catabolic processes, regulating the rate of cell growth and proliferation. Dysregulation of the mTOR signaling pathway occurs frequently in a variety of human tumors, and thus, mTOR has emerged as an important target for the design of anticancer agents. mTOR is found in two distinct multiprotein complexes within cells, mTORC1 and mTORC2. These two complexes consist of unique mTOR-interacting proteins and are regulated by different mechanisms. Enormous advances have been made in the development of drugs known as mTOR inhibitors. Rapamycin, the first defined inhibitor of mTOR, showed effectiveness as an anticancer agent in various preclinical models. Rapamycin analogues (rapalogs) with better pharmacologic properties have been developed. However, the clinical success of rapalogs has been limited to a few types of cancer. The discovery that mTORC2 directly phosphorylates Akt, an important survival kinase, adds new insight into the role of mTORC2 in cancer. This novel finding prompted efforts to develop the second generation of mTOR inhibitors that are able to target both mTORC1 and mTORC2. Here, we review the recent advances in the mTOR field and focus specifically on the current development of the second generation of mTOR inhibitors as anticancer agents.


Assuntos
Humanos , Antineoplásicos , Farmacologia , Proliferação de Células , Furanos , Farmacologia , Imidazóis , Farmacologia , Indóis , Farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Morfolinas , Farmacologia , Complexos Multiproteicos , Naftiridinas , Farmacologia , Neoplasias , Patologia , Fosfatidilinositol 3-Quinases , Metabolismo , Proteínas Proto-Oncogênicas c-akt , Metabolismo , Purinas , Farmacologia , Piridinas , Farmacologia , Pirimidinas , Farmacologia , Quinolinas , Farmacologia , Transdução de Sinais , Sirolimo , Farmacologia , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA