Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2612: 195-224, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36795369

RESUMO

Traditional immunoassays to detect secreted or intracellular proteins can be tedious, require multiple washing steps, and are not easily adaptable to a high-throughput screening (HTS) format. To overcome these limitations, we developed Lumit, a novel immunoassay approach that combines bioluminescent enzyme subunit complementation technology and immunodetection. This bioluminescent immunoassay does not require washes or liquid transfers and takes less than 2 h to complete in a homogeneous "Add and Read" format. In this chapter, we describe step-by-step protocols to create Lumit immunoassays for the detection of (1) secreted cytokines from cells, (2) phosphorylation levels of a specific signaling pathway node protein, and (3) a biochemical protein-protein interaction between a viral surface protein and its human receptor.


Assuntos
Citocinas , Testes Imunológicos , Humanos , Imunoensaio/métodos
2.
Nat Commun ; 11(1): 2743, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32488087

RESUMO

Concerted multidisciplinary efforts have led to the development of Cyclin-Dependent Kinase inhibitors (CDKi's) as small molecule drugs and chemical probes of intracellular CDK function. However, conflicting data has been reported on the inhibitory potency of CDKi's and a systematic characterization of affinity and selectivity against intracellular CDKs is lacking. We have developed a panel of cell-permeable energy transfer probes to quantify target occupancy for all 21 human CDKs in live cells, and present a comprehensive evaluation of intracellular isozyme potency and selectivity for a collection of 46 clinically-advanced CDKi's and tool molecules. We observed unexpected intracellular activity profiles for a number of CDKi's, offering avenues for repurposing of highly potent molecules as probes for previously unreported targets. Overall, we provide a broadly applicable method for evaluating the selectivity of CDK inhibitors in living cells, and present a refined set of tool molecules to study CDK function.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteína Quinase CDC2 , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Quinase 9 Dependente de Ciclina , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Fosforilação , Relação Estrutura-Atividade
3.
Commun Biol ; 3: 8, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31909200

RESUMO

Monitoring cellular signaling events can help better understand cell behavior in health and disease. Traditional immunoassays to study proteins involved in signaling can be tedious, require multiple steps, and are not easily adaptable to high throughput screening (HTS). Here, we describe a new immunoassay approach based on bioluminescent enzyme complementation. This immunoassay takes less than two hours to complete in a homogeneous "Add and Read" format and was successfully used to monitor multiple signaling pathways' activation through specific nodes of phosphorylation (e.g pIκBα, pAKT, and pSTAT3). We also tested deactivation of these pathways with small and large molecule inhibitors and obtained the expected pharmacology. This approach does not require cell engineering. Therefore, the phosphorylation of an endogenous substrate is detected in any cell type. Our results demonstrate that this technology can be broadly adapted to streamline the analysis of signaling pathways of interest or the identification of pathway specific inhibitors.


Assuntos
Imunoensaio/métodos , Medições Luminescentes/métodos , Transdução de Sinais , Biologia Celular/instrumentação , Descoberta de Drogas/instrumentação , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo
4.
Mol Cancer Res ; 15(3): 348-357, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28031410

RESUMO

JAKs are non-receptor tyrosine kinases that are generally found in association with cytokine receptors. In the canonical pathway, roles of JAKs have well been established in activating STATs in response to cytokine stimulation to modulate gene transcription. In contrast, a noncanonical role of JAK2 has recently been discovered, in which JAK2 in the nucleus imparts the epigenetic regulation of gene transcription through phosphorylation of tyrosine 41 on the histone protein H3. Recent work further demonstrated that this noncanonical mechanism is conserved with JAK1, which is activated by the autocrine cytokines IL6 and IL10 in activated B-cell-like diffuse large B-cell lymphoma (ABC DLBCL), a cancer type that is particularly difficult to treat and has poor prognosis. However, how JAK1 gains access to the nucleus to enable epigenetic regulation remains undefined. Here, we investigated this question and revealed that JAK1 has a classical nuclear localization signal toward the N-terminal region, which can be recognized by multiple importin α isoforms. Moreover, the nuclear import of JAK1 is independent of its kinase activity but is required for the optimal expansion of ABC DLBCL cells in vitroImplications: This study demonstrates that the nuclear import of JAK1 is essential for the optimal fitness of ABC DLBCL cells, and targeting JAK1 nuclear localization is a potential therapeutic strategy for ABC DLBCL. Mol Cancer Res; 15(3); 348-57. ©2016 AACR.


Assuntos
Núcleo Celular/metabolismo , Janus Quinase 1/metabolismo , Linfoma Difuso de Grandes Células B/metabolismo , Sinais de Localização Nuclear/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Sobrevivência Celular/fisiologia , Humanos , Janus Quinase 1/genética , Linfoma Difuso de Grandes Células B/enzimologia , Linfoma Difuso de Grandes Células B/genética , Fosforilação , Transdução de Sinais
5.
J Biol Chem ; 290(29): 17967-17984, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26060253

RESUMO

Activation of IκB kinase (IKK) and NF-κB by genotoxic stresses modulates apoptotic responses and production of inflammatory mediators, thereby contributing to therapy resistance and premature aging. We previously reported that genotoxic agents induce nuclear localization of NF-κB essential modulator (NEMO) via an undefined mechanism to arbitrate subsequent DNA damage-dependent IKK/NF-κB signaling. Here we show that a nonclassical nuclear import pathway via IPO3 (importin 3, transportin 2) mediates stress-induced NEMO nuclear translocation. We found putative nuclear localization signals in NEMO whose mutations disrupted stress-inducible nuclear translocation of NEMO and IKK/NF-κB activation in stably reconstituted NEMO-deficient cells. RNAi screening of both importin α and ß family members, as well as co-immunoprecipitation analyses, revealed that a nonclassical importin ß family member, IPO3, was the only importin that was able to associate with NEMO and whose reduced expression prevented genotoxic stress-induced NEMO nuclear translocation, IKK/NF-κB activation, and inflammatory cytokine transcription. Recombinant IPO3 interacted with recombinant NEMO but not the nuclear localization signal mutant version and induced nuclear import of NEMO in digitonin-permeabilized cells. We also provide evidence that NEMO is disengaged from IKK complex following genotoxic stress induction. Thus, the IPO3 nuclear import pathway is an early and crucial determinant of the IKK/NF-κB signaling arm of the mammalian DNA damage response.


Assuntos
Dano ao DNA , Quinase I-kappa B/metabolismo , NF-kappa B/imunologia , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Linhagem Celular , Células HEK293 , Células HeLa , Humanos , Quinase I-kappa B/química , Quinase I-kappa B/imunologia , Camundongos , Dados de Sequência Molecular , Sinais de Localização Nuclear , beta Carioferinas/imunologia
6.
PLoS One ; 10(3): e0116374, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25742655

RESUMO

NF-κB essential modulator, NEMO, plays a key role in canonical NF-κB signaling induced by a variety of stimuli, including cytokines and genotoxic agents. To dissect the different biochemical and functional roles of NEMO in NF-κB signaling, various mutant forms of NEMO have been previously analyzed. However, transient or stable overexpression of wild-type NEMO can significantly inhibit NF-κB activation, thereby confounding the analysis of NEMO mutant phenotypes. What levels of NEMO overexpression lead to such an artifact and what levels are tolerated with no significant impact on NEMO function in NF-κB activation are currently unknown. Here we purified full-length recombinant human NEMO protein and used it as a standard to quantify the average number of NEMO molecules per cell in a 1.3E2 NEMO-deficient murine pre-B cell clone stably reconstituted with full-length human NEMO (C5). We determined that the C5 cell clone has an average of 4 x 10(5) molecules of NEMO per cell. Stable reconstitution of 1.3E2 cells with different numbers of NEMO molecules per cell has demonstrated that a 10-fold range of NEMO expression (0.6-6x10(5) molecules per cell) yields statistically equivalent NF-κB activation in response to the DNA damaging agent etoposide. Using the C5 cell line, we also quantified the number of NEMO molecules per cell in several commonly employed human cell lines. These results establish baseline numbers of endogenous NEMO per cell and highlight surprisingly normal functionality of NEMO in the DNA damage pathway over a wide range of expression levels that can provide a guideline for future NEMO reconstitution studies.


Assuntos
Dano ao DNA/fisiologia , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Células Precursoras de Linfócitos B/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Transdução de Sinais/fisiologia
7.
Biochem J ; 464(1): 35-48, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25145386

RESUMO

MondoA is a basic helix-loop-helix (bHLH)/leucine zipper (ZIP) transcription factor that is expressed predominantly in skeletal muscle. Studies in vitro suggest that the Max-like protein X (MondoA:Mlx) heterodimer senses the intracellular energy status and directly targets the promoter region of thioredoxin interacting protein (Txnip) and possibly glycolytic enzymes. We generated MondoA-inactivated (MondoA-/-) mice by gene targeting. MondoA-/- mice had normal body weight at birth, exhibited normal growth and appeared to be healthy. However, they exhibited unique metabolic characteristics. MondoA-/- mice built up serum lactate and alanine levels and utilized fatty acids for fuel during exercise. Gene expression and promoter analysis suggested that MondoA functionally represses peroxisome-proliferator-activated receptor γ co-activator-1α (PGC-1α)-mediated activation of pyruvate dehydrogenase kinase 4 (PDK-4) transcription. PDK4 normally down-regulates the activity of pyruvate dehydrogenase, an enzyme complex that catalyses the decarboxylation of pyruvate to acetyl-CoA for entry into the Krebs cycle; in the absence of MondoA, pyruvate is diverted towards lactate and alanine, both products of glycolysis. Dynamic testing revealed that MondoA-/- mice excel in sprinting as their skeletal muscles display an enhanced glycolytic capacity. Our studies uncover a hitherto unappreciated function of MondoA in fuel selection in vivo. Lack of MondoA results in enhanced exercise capacity with sprinting.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/deficiência , Músculo Esquelético/metabolismo , Condicionamento Físico Animal/fisiologia , Resistência Física/fisiologia , Animais , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Condicionamento Físico Animal/métodos
8.
FEBS J ; 279(10): 1883-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22429297

RESUMO

Although improving glucose metabolism by inhibition of pyruvate dehydrogenase kinase 4 (PDK4) may prove beneficial in the treatment of type 2 diabetes or diet-induced obesity, it may have detrimental effects by inhibiting fatty acid oxidation. Peroxisome proliferator-activated receptor α (PPARα) agonists are often used to treat dyslipidemia in patients, especially in type 2 diabetes. Combinational treatment using a PDK4 inhibitor and PPARα agonists may prove beneficial. However, PPARα agonists may be less effective in the presence of a PDK4 inhibitor because PPARα agonists induce PDK4 expression. In the present study, the effects of clofibric acid, a PPARα agonist, on blood and liver lipids were determined in wild-type and PDK4 knockout mice fed a high-fat diet. As expected, treatment of wild-type mice with clofibric acid resulted in less body weight gain, smaller epididymal fat pads, greater insulin sensitivity, and lower levels of serum and liver triacylglycerol. Surprisingly, rather than decreasing the effectiveness of clofibric acid, PDK4 deficiency enhanced the beneficial effects of clofibric acid on hepatic steatosis, reduced blood glucose levels, and did not prevent the positive effects of clofibric acid on serum triacylglycerols and free fatty acids. The metabolic effects of clofibric acid are therefore independent of the induction of PDK4 expression. The additive beneficial effects on hepatic steatosis may be due to induction of increased capacity for fatty acid oxidation and partial uncoupling of oxidative phosphorylation by clofibric acid, and a reduction in the capacity for fatty acid synthesis as a result of PDK4 deficiency.


Assuntos
Ácido Clofíbrico/farmacologia , Fígado Gorduroso/patologia , Hipolipemiantes/farmacologia , Proteínas Quinases/deficiência , Animais , Ácido Clofíbrico/uso terapêutico , Dieta Hiperlipídica , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Hipolipemiantes/uso terapêutico , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Proteínas Quinases/genética , Proteínas Quinases/metabolismo
9.
Biochem J ; 423(2): 243-52, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19627255

RESUMO

The hypothesis that PDHK4 (pyruvate dehydrogenase kinase isoenzyme 4) has potential as a target for the treatment of type 2 diabetes was tested by feeding wild-type and PDHK4 knockout mice a high saturated fat diet that induces hyperglycemia, hyperinsulinaemia, glucose intolerance, hepatic steatosis and obesity. Previous studies have shown that PDHK4 deficiency lowers blood glucose by limiting the supply of three carbon gluconeogenic substrates to the liver. There is concern, however, that the increase in glucose oxidation caused by less inhibition of the pyruvate dehydrogenase complex by phosphorylation will inhibit fatty acid oxidation, promote ectopic fat accumulation and worsen insulin sensitivity. This was examined by feeding wild-type and PDHK4 knockout mice a high saturated fat diet for 8 months. Fasting blood glucose levels increased gradually in both groups but remained significantly lower in the PDHK4 knockout mice. Hyperinsulinaemia developed in both groups, but glucose tolerance was better and body weight was lower in the PDHK4 knockout mice. At termination, less fat was present in the liver and skeletal muscle of the PDHK4 knockout mice. Higher amounts of PGC-1alpha [PPARgamma (peroxisome proliferator-activated receptor gamma) coactivator 1alpha] and PPARalpha and lower amounts of fatty acid synthase and acetyl-CoA carboxylase isoenzyme 1 were present in the liver of the PDHK4 knockout mice. These findings suggest PDHK4 deficiency creates conditions that alter upstream signalling components involved in the regulation of lipid metabolism. The findings support the hypothesis that PDHK4 is a viable target for the treatment of type 2 diabetes.


Assuntos
Citoproteção/genética , Gorduras na Dieta/efeitos adversos , Proteínas Serina-Treonina Quinases/genética , Tecido Adiposo/metabolismo , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Animais , Dieta Aterogênica , Gorduras na Dieta/farmacologia , Ácidos Graxos/efeitos adversos , Ácidos Graxos/farmacologia , Hiperglicemia/genética , Hiperinsulinismo/genética , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piruvato Desidrogenase Quinase de Transferência de Acetil , Fatores de Tempo , Aumento de Peso/genética
10.
Biochem Biophys Res Commun ; 378(3): 584-8, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19061868

RESUMO

Translation of hepatitis C virus (HCV) RNA is initiated by internal loading of the ribosome into the HCV internal ribosome entry site (IRES). Previously, heterogeneous ribonucleoprotein L (hnRNP L) was shown to bind specifically to the 3' border region of the HCV IRES and enhance HCV mRNA translation. Here, we provide evidence for the functional requirement of hnRNP L for the HCV IRES-mediated translation initiation using specific RNA aptamers. In vitro selection techniques were employed to isolate RNA aptamers against hnRNP L, which were shown to contain consensus sequences with repetitive ACAC/U. The hnRNP L-specific RNA aptamers efficiently inhibited the in vitro translation reactions mediated by the HCV IRES in rabbit reticulocyte lysates. RNA ligands with only (ACAU)5 or (AC)10 nucleotide sequences could also specifically bind to hnRNP L, and specifically and effectively impeded in vitro translation reactions controlled by the HCV IRES. Importantly, the hnRNP L-specific RNA aptamers inhibited the HCV IRES function in cells in a dose-dependent manner, and the aptamer-mediated inhibition of the HCV IRES was considerably relieved by the addition of hnRNP L-expressing vector. These results strongly demonstrate the functional requirement of cellular hnRNP L for the HCV IRES activity.


Assuntos
Hepacivirus/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/fisiologia , Biossíntese de Proteínas , RNA Viral/metabolismo , Ribossomos/virologia , Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Sistema Livre de Células , Hepacivirus/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Fígado/virologia , RNA Viral/genética
11.
RNA ; 10(8): 1277-90, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15247433

RESUMO

Hepatitis C virus (HCV)-encoded nonstructural protein 3 (NS3) possesses protease, NTPase, and helicase activities, which are considered essential for viral proliferation. Thus, HCV NS3 is a good putative therapeutic target protein for the development of anti-HCV agents. In this study, we isolated specific RNA aptamers to the helicase domain of HCV NS3 from a combinatorial RNA library with 40-nucleotide random sequences using in vitro selection techniques. The isolated RNAs were observed to very avidly bind the HCV helicase with an apparent Kd of 990 pM in contrast to original pool RNAs with a Kd of >1 microM. These RNA ligands appear to impede binding of substrate RNA to the HCV helicase and can act as potent decoys to competitively inhibit helicase activity with high efficiency compared with poly(U) or tRNA. The minimal binding domain of the ligands was determined to evaluate the structural features of the isolated RNA molecules. Interestingly, part of binding motif of the RNA aptamers consists of similar secondary structure to the 3'-end of HCV negative-strand RNA. Moreover, intracellular NS3 protein can be specifically detected in situ with the RNA aptamers, indicating that the selected RNAs are very specific to the HCV NS3 helicase. Furthermore, the RNA aptamers partially inhibited RNA synthesis of HCV subgenomic replicon in Huh-7 hepatoma cell lines. These results suggest that the RNA aptamers selected in vitro could be useful not only as therapeutic and diagnostic agents of HCV infection but also as a powerful tool for the study of HCV helicase mechanism.


Assuntos
Hepacivirus/metabolismo , Hepatite C/metabolismo , RNA Helicases/metabolismo , RNA/metabolismo , Adenosina Trifosfatases/metabolismo , Sítios de Ligação , Hepacivirus/enzimologia , Hepatite C/enzimologia , Hepatite C/genética , Humanos , Ligação Proteica
12.
J Biol Chem ; 279(29): 30053-9, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15138265

RESUMO

Fanconi anemia (FANC) is a heterogeneous genetic disorder characterized by a hypersensitivity to DNA-damaging agents, chromosomal instability, and defective DNA repair. Eight FANC genes have been identified so far, and five of them (FANCA, -C, -E, -F, and -G) assemble in a multinuclear complex and function at least in part in a complex to activate FANCD2 by monoubiquitination. Here we show that FANCA and FANCG are redox-sensitive proteins that are multimerized and/or form a nuclear complex in response to oxidative stress/damage. Both FANCA and FANCG proteins exist as monomers under non-oxidizing conditions, whereas they become multimers following H2O2 treatment. Treatment of cells with oxidizing agent not only triggers the multimeric complex of FANCA and FANCG in vivo but also induces the interaction between FANCA and FANCG. N-Ethylmaleimide treatment abolishes multimerization and interaction of FANCA and FANCG in vitro. Taken together, our results lead us to conclude that FANCA and FANCG uniquely respond to oxidative damage by forming complex(es) via intermolecular disulfide linkage(s), which may be crucial in forming such complexes and in determining their function.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Anemia de Fanconi/metabolismo , Estresse Oxidativo , Proteínas/fisiologia , Animais , Western Blotting , Células COS , Clonagem Molecular , Dano ao DNA , Reparo do DNA , DNA Complementar/metabolismo , Dimerização , Dissulfetos , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Etilmaleimida/farmacologia , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/farmacologia , Mitomicina/farmacologia , Modelos Biológicos , Oxidantes/farmacologia , Oxirredução , Oxigênio/metabolismo , Testes de Precipitina , Estrutura Terciária de Proteína
13.
FEBS Lett ; 548(1-3): 85-9, 2003 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12885412

RESUMO

Myasthenia gravis (MG) and its animal model, experimental autoimmune MG (EAMG), are mainly caused by autoantibodies directed against acetylcholine receptors (AChR) located in the postsynaptic muscle membrane. Previously, we isolated an RNA aptamer with 2'-fluoropyrimidines using in vitro selection techniques that acted as an effective decoy against both a rat monoclonal antibody called mAb198, which recognizes the main immunogenic region on the AChR, and a significant fraction of patient autoantibodies with MG. To investigate the therapeutic potential of the RNA, we tested the ability of the RNA aptamer to protect the receptors in vivo from mAb198. Clinical symptoms of EAMG in rats engendered by passive transfer of mAb198 were efficiently inhibited by a truncated RNA aptamer that was modified with polyethylene glycol, but not by control scrambled RNA. Moreover, the loss of AChR in the animals induced by the antibody was also significantly blocked with the modified RNA aptamer. These results suggested that RNA aptamers could be applied for antigen-specific treatment for autoimmune diseases including MG.


Assuntos
Miastenia Gravis Autoimune Experimental/prevenção & controle , RNA/farmacologia , RNA/uso terapêutico , Receptores Colinérgicos/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Autoanticorpos/metabolismo , Autoanticorpos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Humanos , Miastenia Gravis Autoimune Experimental/imunologia , Polietilenoglicóis , RNA/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/análise
14.
Biochem Biophys Res Commun ; 290(2): 656-62, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11785949

RESUMO

Myasthenia gravis (MG) is mainly engendered by autoantibodies directed against acetylcholine receptors (AChRs) located in the postsynaptic muscle cell membrane. Previously, we isolated an RNA aptamer with 2'-amino pyrimidines using in vitro selection techniques that acted as a decoy against both a rat monoclonal antibody called mAb198, which recognizes the main immunogenic region on the AChR, and patient autoantibodies with MG (1). However, low affinity of this RNA to mAb198 relative to that of AChR might limit potential of the RNA as an inhibitor of the autoantibodies. To improve decoy activity of the RNA aptamer against autoantibodies, here we employed in vitro selection methods with RNA libraries containing extra random nucleotides extended to the 3' end of previously selected RNA sequences. RNAs isolated in this study showed significant increases in the binding affinities to mAb198 as well as bioactivities protecting AChRs on human cells from both mAb198 and patient autoantibodies, compared with the previous RNA aptamers. These results have important implications for the development of antigen-specific modulation of autoimmune diseases including MG.


Assuntos
Autoanticorpos/efeitos dos fármacos , Ligação Competitiva/efeitos dos fármacos , Miastenia Gravis/imunologia , RNA/farmacologia , Receptores Colinérgicos/imunologia , Animais , Anticorpos Monoclonais/efeitos dos fármacos , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos/imunologia , Autoanticorpos/metabolismo , Ligação Competitiva/imunologia , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Epitopos/imunologia , Epitopos/metabolismo , Humanos , Miastenia Gravis/terapia , Conformação de Ácido Nucleico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , RNA/isolamento & purificação , RNA/metabolismo , Ratos , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...