Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Mediators Inflamm ; 2021: 1180543, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34720748

RESUMO

The Tn antigen, an N-acetylgalactosamine structure linked to serine or threonine, has been shown to induce high-specificity, high-affinity anti-Tn antibodies in mice. Maternal immunization with the Tn vaccine increases serum anti-Tn antibody titers and attenuates hyperoxia-induced kidney injury in neonatal rats. However, immunizing mothers to treat neonatal kidney disease is clinically impractical. This study is aimed at determining whether anti-Tn monoclonal antibody treatment ameliorates hyperoxia-induced kidney injury in neonatal mice. Newborn BALB/c mice were exposed to room air (RA) or normobaric hyperoxia (85% O2) for 1 week. On postnatal days 2, 4, and 6, the mice were injected intraperitoneally with PBS alone or with anti-Tn monoclonal antibodies at 25 µg/g body weight in 50 µL phosphate-buffered saline (PBS). The mice were divided into four study groups: RA + PBS, RA + anti-Tn monoclonal antibody, O2 + PBS, and O2 + anti-Tn monoclonal antibody. The kidneys were excised for histology, oxidative stress, cytokine, and Western blot analyses on postnatal day 7. The O2 + PBS mice exhibited significantly higher kidney injury scores, 8-hydroxy-2'-deoxyguanosine (8-OHdG) and nuclear factor-κB (NF-κB) expression, and cytokine levels than did the RA + PBS mice or RA + anti-Tn mice. Anti-Tn monoclonal antibody treatment reduced kidney injury and cytokine levels to normoxic levels. The attenuation of kidney injury was accompanied by a reduction of oxidative stress and NF-κB expression. Therefore, we propose that anti-Tn monoclonal antibody treatment ameliorates hyperoxia-induced kidney injury by suppressing oxidative stress and inflammation in neonatal mice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos Glicosídicos Associados a Tumores/imunologia , Hiperóxia/complicações , Inflamação/prevenção & controle , Rim/patologia , Estresse Oxidativo , Animais , Animais Recém-Nascidos , Citocinas/análise , Feminino , Quinase I-kappa B/análise , Rim/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator de Transcrição RelA/análise
2.
J Intern Med ; 285(3): 301-316, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30644612

RESUMO

AIMS: There are limited data on the role of human monocyte subsets in ST-elevation myocardial infarction (STEMI). The study aimed to establish the relationship between monocyte subsets, their phagocytic and nuclear factor κB (NFκB) activity and outcomes in STEMI. METHODS: Monocyte subsets and their phagocytic activity and intracellular levels of inhibitory κB kinase ß (IKKß, marker of NFκB activity) were measured by flow cytometry in 245 patients with STEMI, median follow-up of 46 months. RESULTS: Mon2 (CD14++CD16+CCR2+) counts were independently predictive of major adverse cardiovascular events (MACE) [4th quartile HR 3.42 (95% CI 1.43-8.16), P = 0.006 and 3rd quartile HR 2.88 (95% CI 1.19-7.00), P = 0.02 vs. 1st quartile]. Mon2 subset was the only subset associated with higher occurrence of heart failure (4th quartile vs. 1st quartile, sevenfold, P = 0.001 on univariate analysis; fivefold, P = 0.04 on multivariable analysis). On receiver operating characteristic, analysis including of Mon2 improved prognostic value of troponin T and creatine kinase for MACE and heart failure (HF). Higher intracellular Mon2 IKKß levels were associated with 10-fold lower occurrence of HF on multivariable analysis (4th vs. 1st quartiles, P = 0.03). Abnormal Mon1 and Mon2 phagocytic capacities were related to HF development, but the association was dependent on the infarct size and other prognosticators. High Mon2 levels were associated with lower ejection fraction after STEMI onset (P = 0.001) and at 6-month follow-up (P < 0.001). CONCLUSIONS: Abnormal Mon2 characteristics have a unique association with poor outcome in patients with STEMI. The relation of Mon2 with occurrence of HF is strongly and independently related to their functional status, which may have potential therapeutic implications.


Assuntos
Insuficiência Cardíaca , Quinase I-kappa B , Monócitos , NF-kappa B , Infarto do Miocárdio com Supradesnível do Segmento ST , Biomarcadores/análise , Biomarcadores/metabolismo , Contagem de Células/métodos , Correlação de Dados , Feminino , Citometria de Fluxo , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/etiologia , Humanos , Quinase I-kappa B/análise , Quinase I-kappa B/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/classificação , Monócitos/fisiologia , NF-kappa B/análise , NF-kappa B/metabolismo , Avaliação de Resultados em Cuidados de Saúde , Fagocitose , Prognóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/sangue , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Volume Sistólico
3.
São Paulo; s.n; s.n; 2019. 113 p. ilus, tab, graf.
Tese em Português | LILACS | ID: biblio-1015182

RESUMO

O câncer de pulmão é o tipo de câncer que apresenta o maior índice de mortalidade em todo o mundo. As alterações genéticas mais frequentes em câncer de pulmão são as mutações pontuais no oncogene que codifica a GTPase KRAS. Apesar destas mutações estarem diretamente ligadas à oncogênese, terapias que visam inibir diretamente a proteína Ras falharam em ensaios clínicos. Uma das propriedades mais importantes na oncogênese é a aquisição de capacidade metastática tumoral. Desta forma, o objetivo deste projeto é identificar alvos terapêuticos que inibam as metástases tumorais induzidas pelo oncogene KRAS no pulmão. Com base em relatos recentes mostrando que a forma oncogênica de KRAS promove, não só a iniciação tumoral, mas também promove a aquisição de um fenótipo metastático, a hipótese deste projeto é que (1) a capacidade mestastática tumoral induzida por KRAS no pulmão é potencializada pela quinase IKKß; e (2) que a inibição desta quinase reduzirá a capacidade invasiva celular e metastática tumoral. Esta hipótese foi formulada com base em estudos anteriores, os quais demonstraram que o principal substrato da IKKß, o fator de transcrição NF-κB, é ativado por KRAS em tumores pulmonares in situ de forma dependente da IKKß, que o NF-κB é capaz de promover metástase em diferentes modelos tumorais, e que a inibição da atividade da IKKß com um inibidor farmacológico em um modelo animal de câncer de pulmão induzido por KRAS, diminui o crescimento tumoral e a progressão tumoral para graus histológicos mais avançados. Nosso objetivo era avaliar se a inibição de IKKß é capaz de afetar a migração e invasão de células portadoras de mutação em KRAS in vitro e se a inibição de IKKß é capaz de afetar a capacidade metatática dessas células in vivo. Primeiramente, avaliamos a expressão de enzimas relacionadas ao fenótipo metastático, as metaloproteinases de matriz 2 e 9 (MMP-2 e MMP-9) e, também uma molécula intimamente relacionada ao processo de adesão mediado por integrinas, FAK (quinase de adesão focal), frente a inibição de IKKß através de um inibidor farmacológico altamente especifico (Composto A) e frente a inibição genética de IKKß por interferência de RNA (siRNA) em células A549 e H358. Avaliamos também a atividade das MMPs frente inibição genética de KRAS (siKRAS) e IKKß (siIKKß) e vimos que IKKß parece modular a expressão ou atividade de MMP-9 e reduz a expressão de FAK. Já a expressão de MMP-2 não apresentou alteração. Posteriormente avaliamos migração na célula A549 e invasão nas células A549 e H358 com inibição de IKKß, por ensaios Transwell, e observamos uma redução da migração e invasão celular in vitro. Em seguida, fomos gerar linhagens celulares paraa expressar luciferase, as linhagens A549 pLUC e H358 pLUC. Os clones A549 pLUC B4 e H358 pLUC F1 com inibição de KRAS e IKKß por interferência de RNA, foram injetados pela veia da cauda nesses camundongos e as metástases foram monitoradas por imageamento in vivo. Houve metástases em 20% dos animais com siIKKß na região anatômica da boca. Os animais que receberam siControle e siKRAS não apresentaram nenhuma metástase visível no equipamento, mas foi observado micrometástases nas análises histológicas dos pulmões. O resultado do experimento de metástase in vivo é inesperado, não só pelo fato de ocorrer no grupo experimental siIKKß, mas também pelo local anatômico do tumor, sendo necessária uma maior investigação do papel de IKKß nesse processo, podendo ser um resultado aleatório. Quando avaliamos em conjunto, nossos resultados sugerem que a quinase IKKß desempenha um papel importante no fenótipo migratório e invasivo de células pulmonares portadoras de KRAS oncogênica, contribuindo para a capacidade metastática


Lung cancer is the leading cause of cancer deaths worldwide. The most frequent genetic changes found in lung cancer are driver mutations in the KRAS proto-oncogene. Even though KRAS mutations have been causally linked to the oncogenic process, therapies targeted to oncogenic RAS have failed in clinical trials. One of the main characteristics in oncogenesis is the ability of tumors to acquire metastatic capability. The objective of this project is to identify therapeutic targets that reduce KRASinduced lung cancer metastasis. Based on previous reports that oncogenic KRAS, drives not only tumor initiation, but also promotes a metastatic phenotype, the hypothesis of this project is that (1) the acquisition of metastatic ability induced by KRAS in the lung is potentiated by the IKK kinase; and (2) that IKKß inhibition will reduce KRAS-induced cell invasive properties and KRAS-induced tumor metastasis. This hypothesis has been formulated on the basis of previous studies showing that the main IKKß substrate, the transcription factor NF-κB, is activated by KRAS in lung tumors in situ in an IKKß-dependent manner, that NF-κB is known to promote metastasis in different tumor models, and that pharmacological IKKß inhibition in a KRAS-induced lung cancer mouse model reduces tumor growth and progression to higher histological tumor grades. Our goal was evaluate how inhibition of IKKß affects migration and invasion of KRAS-positive lung cells in vitro and whether inhibition of IKKß is capable of affecting the metatactic capacity of these cells in vivo. First, we evaluated the expression of enzymes involved in the metastatic phenotype, matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) and also a molecule involved in the integrinmediated adhesion, FAK (focal adhesion kinase), we targeted IKKß by a highly specific IKK inhibitor (Compound A) or with RNA interference in A549 and H358 cells. We also used colorimetric Matrix Biotrak Activity Assay System to measure the activity of MMPs with RNA interference for KRAS (siKRAS) and IKKß (IKKß) and we have seen that IKKß appears to modulate the expression or activity of MMP-9 and decreases the expression of FAK. The expression of MMP-2 did not change. Then we evaluated migration in A549 cell and invasion in A549 and H358 cells with inhibition of IKK by RNA interference or with Compound A treatment in Transwell assays, and observed a significantly reduced cell migration and invasion in vitro. We then generated cell lines to express luciferase, the A549 pLUC and H358 pLUC lines. A549 pLUC B4 and H358 pLUC F1 cells with RNA interference for KRAS and IKKß were injected in the tail vein in nude (balb/c) mice and metastases were monitored by in vivo imaging. There were metastases in 20% of IKKß animals in the anatomical region of the mouth. Animals that received siControl and siKRAS had no visible metastasis in the live imaging, but micrometastases were observed in the histological analyzes of the lungs. The result of this experiment is unexpected, not only due to the fact that it occurs in the IKKß experimental group, but also due to the anatomical site of the tumor, and a further investigation of the role of IKKß in this process, can be a random result. When evaluated together, our results suggest that the IKKß kinase plays an important role in the migratory and invasive phenotype of in KRAS positive lung cancer cells, contributing to metastatic capacity


Assuntos
Animais , Masculino , Feminino , Camundongos , Quinase I-kappa B/análise , Neoplasias Pulmonares/tratamento farmacológico , Metástase Neoplásica , Técnicas In Vitro , Western Blotting/instrumentação , Proteínas ras/classificação , Inibidores de Proteínas Quinases
4.
Urol Int ; 101(2): 190-196, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30089311

RESUMO

BACKGROUND: In the United Kingdom, 8,000 cases of renal cancer are diagnosed each year, with a 5-year survival rate of 50%. Treatment options are limited; a potential therapeutic target is the non-canonical nuclear factor-kappa B (NF-κB) pathway. This pathway plays a role in multiple oncogenic processes in solid tumors. The aim of this study was to investigate the non-canonical nuclear factor pathway in renal cell carcinoma (RCC). MATERIALS AND METHODS: NIK, IKKα, and RelB were investigated via immunohistochemistry in a cohort of 192 patients with clear cell renal cancer. RESULTS: High cytoplasmic NIK was associated with poorer cancer-specific survival (p = 0.006) and 10-year survival stratified from 85% (low) to 65% (high, p = 0.005). Similarly, high cytoplasmic RelB was associated with poorer cancer-specific survival (p = 0.041) and 10-year survival stratified from 88% (low) to 73% (high, p = 0.030). When clinicopathological characteristics were assessed, cytoplasmic NIK was associated with survival (p = 0.014), whereas cytoplasmic RelB was associated with increased tumor grade (p = 0.020) and decreased inflammation (p = 0.019). Upon multivariate analysis, it was found that cytoplasmic NIK was independently associated with cancer-specific survival (p = 0.009). CONCLUSIONS: The non-canonical NF-κB pathway is associated with poorer cancer-specific survival in RCC patients, making it a viable target for therapeutic intervention. Furthermore, cytoplasmic NIK is a potential prognostic biomarker for this disease.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Renais/química , Quinase I-kappa B/análise , Neoplasias Renais/química , Proteínas Serina-Treonina Quinases/análise , Fator de Transcrição RelB/análise , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/cirurgia , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Nefrectomia , Intervalo Livre de Progressão , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Quinase Induzida por NF-kappaB
5.
Clin Transl Oncol ; 20(12): 1592-1603, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29796997

RESUMO

PURPOSE: Uveal melanoma, although a rare form of cancer, is the most common primary malignancy of the eye in adults. Nuclear factor-κB (NF-κB) is a transcription factor that transactivates genes involved in the regulation of cell growth, apoptosis, angiogenesis, and metastasis, but the molecular mechanisms that negatively regulate NF-κB activation are not fully understood. NF-κB can also be activated by DNA damage pathway through NEMO protein. Therefore, the objective of this study is to elucidate the role of NEMO/IKKγ protein in uveal melanoma patients. METHODS: Seventy-five formalin-fixed paraffin-embedded prospective tissues of uveal melanoma were included in the present study. These cases were reviewed and investigated for the expression of NEMO/IKKγ protein by immunohistochemistry and validated by western blotting along with the qRT-PCR for mRNA expression. Expression levels were correlated with the clinicopathological parameters and patients' outcome. RESULTS: Immunohistochemistry showed cytoplasmic expression of NEMO/IKKγ expression in only 22 out of 75 (29.33%) cases. This result was confirmed by western blotting, and correlated well with the immunohistochemical expression of NEMO/IKKγ protein (48 kDa). In addition, downregulation of this gene was found in 87.93% of the cases when compared with the normal tissues. On statistical analysis, loss of NEMO/IKKγ protein was correlated with neovascularization, high mitotic count, and presence of vascular loop (p < 0.05). There was less overall survival rate with low expression of NEMO/IKKγ protein in patients with uveal melanoma. CONCLUSION: This was the first study suggesting the relevant role of NEMO/IKKγ protein, and highlights the prognostic significance with outcome in uveal melanoma patients. This protein might be used as a screening biomarker in these patients after large-scale validation and translational studies.


Assuntos
Biomarcadores Tumorais/análise , Quinase I-kappa B/biossíntese , Melanoma/patologia , Neoplasias Uveais/patologia , Adulto , Idoso , Feminino , Humanos , Quinase I-kappa B/análise , Masculino , Melanoma/metabolismo , Melanoma/mortalidade , Pessoa de Meia-Idade , Prognóstico , Neoplasias Uveais/metabolismo , Neoplasias Uveais/mortalidade
6.
JCI Insight ; 3(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29367460

RESUMO

Mesenchymal stem cells (MSCs) can give rise to both adipocytes and osteoblasts, but the molecular mechanisms underlying MSC fate determination remain poorly understood. IκB kinase ß (IKKß), a central coordinator of inflammation and immune responses through activation of NF-κB, has been implicated as a critical molecular link between obesity and metabolic disorders. Here, we show that IKKß can reciprocally regulate adipocyte and osteoblast differentiation of murine and human MSCs through an NF-κB-independent mechanism. IKKß is a ß-catenin kinase that phosphorylates the conserved degron motif of ß-catenin to prime it for ß-TrCP-mediated ubiquitination and degradation, thereby increasing adipogenesis and inhibiting osteogenesis in MSCs. Animal studies demonstrated that deficiency of IKKß in BM mesenchymal stromal cells increased bone mass and decreased BM adipocyte formation in adult mice. In humans, IKKß expression in adipose tissue was also positively associated with increased adiposity and elevated ß-catenin phosphorylation. These findings suggest IKKß as a key molecular switch that regulates MSC fate, and they provide potentially novel mechanistic insights into the understanding of the cross-regulation between the evolutionarily conserved IKKß and Wnt/ß-catenin signaling pathways. The IKKß-Wnt axis we uncovered may also have important implications for development, homeostasis, and disease pathogenesis.


Assuntos
Diferenciação Celular/fisiologia , Quinase I-kappa B/metabolismo , Células-Tronco Mesenquimais/metabolismo , Obesidade/patologia , beta Catenina/metabolismo , Gordura Abdominal/patologia , Adipócitos/fisiologia , Adipogenia/fisiologia , Adulto , Animais , Biópsia , Células Cultivadas , Feminino , Humanos , Quinase I-kappa B/análise , Quinase I-kappa B/genética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Modelos Animais , Obesidade/sangue , Osteoblastos/fisiologia , Osteogênese/fisiologia , Fosforilação/fisiologia , Cultura Primária de Células , Proteólise , Ubiquitinação/fisiologia , Via de Sinalização Wnt/fisiologia , Proteínas Contendo Repetições de beta-Transducina/metabolismo
7.
São Paulo; s.n; s.n; 2018. 133 p. graf, tab.
Tese em Português | LILACS | ID: biblio-999085

RESUMO

As alterações genéticas mais frequentes em câncer de pulmão são mutações pontuais que ativam o oncogene KRAS. Embora estas mutações estejam causalmente relacionadas à oncogênese, até hoje diferentes abordagens para inibir as proteínas RAS diretamente não obtiveram sucesso. Portanto, para que melhores alvos terapêuticos para o câncer de pulmão se tornem disponíveis é necessário identificar os mecanismos moleculares ativados por KRAS que estão diretamente envolvidos com a aquisição de propriedades malignas importantes, como o desenvolvimento e a manutenção de um fenótipo tronco-tumoral pelas células iniciadoras de tumor (CITs). CITs, também conhecidas como células tronco-tumorais, são definidas como uma subpopulação de células tumorais capazes de se autorrenovar, iniciar a formação de tumores e sustentar o crescimento tumoral. O desenvolvimento de estratégias terapêuticas dirigidas a estas células é imprescindível para melhorar a eficácia da terapia antitumoral. Uma vez que KRAS está associada a manutenção de um fenótipo tronco-tumoral e ativa o fator de transcrição NF-kB através da quinase IKKß para promover a tumorigênese pulmonar, nós hipotetizamos que a quinase IKKß contribui para o fenótipo tronco-tumoral induzido por KRAS em câncer de pulmão. Nós utilizamos ensaios de formação de tumoresferas para enriquecer e avaliar a função de CITs das linhagens pulmonares positivas para KRAS A549 e H358. As células A549 e H358 formaram tumoresferas em cultura de baixa aderência e, quando comparadas às células derivadas da cultura aderente, as células oriundas da cultura de tumoresferas apresentaram maior crescimento clonogênico, maior expressão de genes associados ao fenótipo tronco por qPCR e maior atividade da quinase IKKß. A inibição da atividade de IKKß através de um inibidor farmacológico altamente específico (Composto A) diminuiu levemente a proliferação de células A549 e H358, sem resultar em morte celular significativa. Entretanto, a inibição da atividade ou da expressão de IKKß por interferência de RNA reduziu a expressão de genes associados ao fenótipo tronco e diminuiu a formação de tumoresferas. A inibição da expressão de IKKß em células A549 reduziu também a capacidade de autorrenovação de CITs. Estes resultados sugerem que IKKß desempenha um papel importante na manutenção do fenótipo tronco-tumoral de CITs pulmonares induzidas por KRAS. Em seguida, nós demonstramos que a inibição da atividade de IKKß afetou preferencialmente a proliferação celular e o crescimento clonogênico de células oriundas da cultura de tumoresfera, sugerindo que IKKß desempenha um papel mais importante em CITs do que em células derivadas da cultura aderente. A análise por citometria de fluxo identificou que células derivadas da cultura de tumoresfera apresentam um enriquecimento para células CD24+ na linhagem A549 e células CD44+ na linhagem H358, sugerindo que estes possam ser marcadores promissores para purificação de CITs nestas linhagens. Adicionalmente, demonstramos, por ensaios de wound-healing de células A549 e H358, que a inibição da atividade de IKKß reduziu a migração celular, uma outra uma propriedade aumentada em CITs. Além disso, mostramos que a atividade da quinase IKKß em células A549 e H358 não depende das vias da MAPK ou PI3K/Akt. Interessantemente, a inibição combinada de IKK (um efetor downstream de KRAS) e de EGFR/ERRB2 (reguladores upstream de KRAS que ativam as vias MAPK e PI3K/Akt) reduziu de forma aditiva a formação de tumoresferas, proliferação e migração celular. Quando avaliados em conjunto, nossos resultados sugerem que a quinase IKKß desempenha um papel importante na biologia de CITs pulmonares portadoras de KRAS oncogênica e que a inibição desta quinase sozinha ou em combinação com a inibição de outras vias pode representar uma estratégia terapêutica promissora a ser explorada para reduzir a recidiva e metástase no câncer de pulmão induzido por KRAS


The most frequent genetic alterations in lung cancer are point mutations that activate the KRAS oncogene. Although these mutations are causally related to oncogenesis, different approaches to inhibit RAS proteins directly have not been successful to date. Therefore, for better therapeutic targets for lung cancer to become available, it is necessary to identify the molecular mechanisms activated by KRAS that are directly involved with important malignant features, such as the development and maintenance of a cancer stem-like phenotype by the tumour-initiating cells (TICs). TICs, also known as cancer stem cells, are defined as a subpopulation of tumour cells able to self-renew, promote tumour initiation, and sustain tumour growth. The development of therapeutic strategies to target these cells is imperative to improve the efficacy of antitumor therapy. Since KRAS is associated with the maintenance of a cancer stem-like phenotype and activates the transcription factor NF-kB through the IKKß kinase to promote lung tumourigenesis, we hypothesised that IKKß kinase contributes to the cancer stem-like phenotype induced by KRAS in lung cancer. We used tumoursphere formation assays to enrich and evaluate the function of TICs of KRAS-mutant cell lines A549 and H358. A549 and H358 cells formed tumourspheres in low adhesion culture and, when compared to cells grown in adherent culture, sphere-derived cells displayed increased clonogenic growth, higher expression of stemness genes by qPCR, and increased IKKß kinase activity . Inhibition of IKKß activity through a highly specific pharmacological inhibitor (Compound A) slightly decreased proliferation of A549 and H358 cells without inducing significant cell death. On the other hand, inhibition of IKKß activity or expression by RNA interference reduced the expression of stemness genes and decreased tumoursphere formation. Inhibition of IKKß expression in A549 cells also reduced TICs self-renewal . These results suggest that IKKß plays an important role in maintaining the cancer stem-like phenotype of KRAS-driven lung TICs. Next, we demonstrated that IKKß inhibition preferentially reduced cell proliferation and clonogenic growth of sphere-derived cells, suggesting that IKKß plays a more important role in TICs than in adherent culture-derived cells. Flow cytometry analysis identified that sphere-derived cells display an enrichment for the surface marker CD24 in A549 cells and CD44 in H358 cells, indicating that these could be promising markers for the purification of TICs in these cell lines. Furthermore, we have shown by wound-healing assays of A549 and H358 cells that IKKß inhibition reduced cell migration , another feature increased in TICs. In addition, we have shown that IKKß activity in A549 and H358 cells does not depend on the MAPK or PI3K/Akt pathways. Interestingly, combined inhibition of IKKß (a downstream effector of KRAS) and EGFR/ERBB2 (upstream regulators of KRAS that activate the MAPK and PI3K/Akt pathways) additively reduced tumoursphere formation, cell proliferation and migration. Taken together, our results suggest that IKKß kinase plays an important role in the biology of KRAS-driven lung TICs, and that inhibition of this kinase alone or in combination with inhibition of other signalling pathways may represent a promising therapeutic strategy to be explored in order to reduce tumour recurrence and metastasis in KRAS-driven lung cancer


Assuntos
Genes ras , Quinase I-kappa B/análise , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamento farmacológico
8.
PLoS One ; 12(8): e0181052, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28797077

RESUMO

Myeloid cell and hepatocyte IKKß may mediate the genesis of obesity and insulin resistance in mice fed high fat diet. However, their gender-specific roles in the pathogenesis of non-alcoholic steatohepatitis (NASH) are not known. Here we demonstrate myeloid IKKß deficiency prevents Western diet-induced obesity and visceral adiposity in females but not in males, and attenuates hyperglycemia, global IR, and NASH in both genders. In contrast, all metabolic sequela including NASH are aggravated by hepatocyte IKKß deficiency (IkbkbΔhep) in male but not female mice. Gene profiling identifies sulfotransferase family 1E (Sult1e1), which encodes a sulfotransferase E1 responsible for inactivation of estrogen, as a gene upregulated in NASH in both genders and most conspicuously in male IkbkbΔhep mice having worst NASH and lowest plasma estradiol levels. LXRα is enriched to LXRE on Sult1e1 promoter in male WT and IkbkbΔhep mice with NASH, and a Sult1e1 promoter activity is increased by LXRα and its ligand and augmented by expression of a S32A mutant of IκBα. These results demonstrate striking gender differences in regulation by IKKß of high cholesterol saturated fat diet-induced metabolic changes including NASH and suggest hepatocyte IKKß is protective in male due at least in part to its ability to repress LXR-induced Sult1e1. Our findings also raise a caution for systemic IKK inhibition for the treatment of NASH as it may exacerbate the disease in male patients.


Assuntos
Quinase I-kappa B/genética , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Sulfotransferases/genética , Adiposidade , Animais , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Hiperglicemia/etiologia , Hiperglicemia/genética , Hiperglicemia/patologia , Quinase I-kappa B/análise , Resistência à Insulina/genética , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Fatores Sexuais , Sulfotransferases/análise , Transcriptoma
9.
Virol J ; 14(1): 31, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28202068

RESUMO

BACKGROUND: Infection of glial cells by human neurotropic polyomavirus JC (JCV), the causative agent of the CNS demyelinating disease progressive multifocal leukoencephalopathy (PML), rapidly inflicts damage to cellular DNA. This activates DNA damage response (DDR) signaling including induction of expression of DNA repair factor Rad51. We previously reported that Rad51 co-operates with the transcription factor NF-κB p65 to activate JCV early transcription. Thus Rad51 induction by JCV infection may provide positive feedback for viral activation early in JCV infection. DDR is also known to stimulate NF-κB activity, a phenomenon known as nucleus to cytoplasm or "inside-out" NF-κB signaling, which is initiated by Ataxia telangiectasia mutated (ATM) protein, a serine/threonine kinase recruited and activated by DNA double-strand breaks. Downstream of ATM, there occurs a series of post-translational modifications of NF-κB essential modulator (NEMO), the γ regulatory subunit of inhibitor of NF-κB (IκB) kinase (IKK), resulting in NF-κB activation. METHODS: We analyzed the effects of downstream pathways in the DDR by phosphospecific Western blots and analysis of the subcellular distribution of NEMO by cell fractionation and immunocytochemistry. The role of DDR in JCV infection was analyzed using a small molecule inhibitor of ATM (KU-55933). NEMO sumoylation was investigated by Western and association of ATM and NEMO by immunoprecipitation/Western blots. RESULTS: We show that JCV infection caused phosphorylation and activation of ATM while KU-55933 inhibited JCV replication. JCV infection caused a redistribution of NEMO from cytoplasm to nucleus. Co-expression of JCV large T-antigen and FLAG-tagged NEMO showed the occurrence of sumoylation of NEMO, while co-expression of ATM and FLAG-NEMO demonstrated physical association between ATM and NEMO. CONCLUSIONS: We propose a model where JCV infection induces both overexpression of Rad51 protein and activation of the nucleus to cytoplasm NF-κB signaling pathway, which then act together to enhance JCV gene expression.


Assuntos
Dano ao DNA , Interações Hospedeiro-Patógeno , Vírus JC/crescimento & desenvolvimento , NF-kappa B/metabolismo , Neuroglia/virologia , Transdução de Sinais , Estresse Fisiológico , Western Blotting , Fracionamento Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Quinase I-kappa B/análise , Imuno-Histoquímica , Vírus JC/genética , Modelos Biológicos , Transporte Proteico , Rad51 Recombinase/metabolismo , Transcrição Gênica
11.
Oncotarget ; 6(28): 25499-505, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26317791

RESUMO

The functional role of IKKα in vivo is pretty complicated, largely due to its diverse functions through cell autonomous and non-autonomous manners. In addition, most of the studies on IKKα were derived from animal models, whether these findings hold true in human tumors remain unclear. Here we examined the expression of IKKα in nasopharyngeal carcinoma, which includes non-keratinizing carcinoma and keratinizing squamous cell carcinoma, and lung squamous cell carcinoma with keratinization and non-keratinization. We demonstrated that IKKα expression was almost negative in keratinizing cancer and higher expression of IKKα was found in non-keratinizing cancer, and that IKKα expression correlated with cellular differentiation of tumors in non-keratinizing nasopharyngeal carcinoma. These findings demonstrate that IKKα is diversely expressed in keratinizing and non-keratinizing carcinomas in the same type of cancer.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/enzimologia , Quinase I-kappa B/análise , Queratinas/análise , Neoplasias Pulmonares/enzimologia , Neoplasias Nasofaríngeas/enzimologia , Biomarcadores Tumorais/genética , Carcinoma , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Quinase I-kappa B/genética , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/mortalidade , Neoplasias Nasofaríngeas/patologia , Fenótipo , Prognóstico , Fatores de Tempo
12.
Clin Exp Med ; 15(3): 269-75, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25149277

RESUMO

Cyclin-dependent kinases (CDKs) have emerged as anti-inflammatory targets. The purpose of this study was to explore the therapeutic effects of a selective CDK7 inhibitor, BS-181, on mice with established collagen-induced arthritis (CIA). CIA mice were administered intraperitoneally with BS-181 (10 mg/kg) twice daily for 2 weeks. Control mice received vehicle only. Arthritis severity and joint histopathology were examined. The proinflammatory cytokines and anti-type II collagen antibodies (anti-CII) were determined by ELISA. IkB kinase (IKK)-ß/NF-κB activation in the arthritic joints was assessed by Western blot. The ratio of Th17 cells was determined by flow cytometry. In vitro, splenocytes from mice with established CIA were stimulated with CII in the presence or absence of BS-181 and cytokines were detected. BS-181 treatment reduced the clinical score and histological damage in CIA mice. The serum proinflammatory cytokines (IL-6, IL-1ß and IL-17) and anti-CII IgG2a levels were also decreased by BS-181 administration. Moreover, IKK-ß/NF-κB signaling pathway was inhibited in arthritic joints. BS-181 administration also decreased the ratio of Th17 cells. In addition, CIA splenocytes pretreated with BS-181 produced less proinflammatory cytokines in vitro. These findings indicate that CDK7 inhibition by BS-181 is effective in the treatment of CIA, which might be mediated by suppression of IKK-ß/NF-κB activation and Th17 cell response.


Assuntos
Anti-Inflamatórios/administração & dosagem , Artrite Experimental/patologia , Artrite Experimental/terapia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Animais , Autoanticorpos/sangue , Western Blotting , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Quinase I-kappa B/análise , Masculino , Camundongos Endogâmicos DBA , NF-kappa B/análise , Índice de Gravidade de Doença , Células Th17/imunologia , Resultado do Tratamento , Quinase Ativadora de Quinase Dependente de Ciclina
13.
Int J Oral Sci ; 6(4): 219-26, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25105817

RESUMO

Bacterial biofilms have emerged as potential critical triggers in the pathogenesis of bisphosphonate (BP)-related osteonecrosis of the jaw (ONJ) or BRONJ. BRONJ lesions have shown to be heavily colonized by oral bacteria, most of these difficult to cultivate and presents many clinical challenges. The purpose of this study was to characterize the bacterial diversity in BRONJ lesions and to determine host immune response. We examined tissue specimens from three cohorts (n=30); patients with periodontal disease without a history of BP therapy (Control, n=10), patients with periodontal disease having history of BP therapy but without ONJ (BP, n=5) and patients with BRONJ (BRONJ, n=15). Denaturing gradient gel electrophoresis of polymerase chain reaction (PCR)-amplified 16S rRNA gene fragments revealed less bacterial diversity in BRONJ than BP and Control cohorts. Sequence analysis detected six phyla with predominant affiliation to Firmicutes in BRONJ (71.6%), BP (70.3%) and Control (59.1%). Significant differences (P<0.05) in genera were observed, between Control/BP, Control/BRONJ and BP/BRONJ cohorts. Enzyme-linked immunosorbent assay (ELISA) results indicated that the levels of myeloperoxidase were significantly lower, whereas interleukin-6 and tumor necrosis factor-alpha levels were moderately elevated in BRONJ patients as compared to Controls. PCR array showed significant changes in BRONJ patients with downregulation of host genes, such as nucleotide-binding oligomerization domain containing protein 2, and cathepsin G, the key modulators for antibacterial response and upregulation of secretory leukocyte protease inhibitor, proteinase 3 and conserved helix-loop-helix ubiquitous kinase. The results suggest that colonization of unique bacterial communities coupled with deficient innate immune response is likely to impact the pathogenesis of ONJ.


Assuntos
Biofilmes , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Boca/microbiologia , Actinobacteria/classificação , Bactérias/classificação , Bacteroidetes/classificação , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/imunologia , Conservadores da Densidade Óssea/uso terapêutico , Catepsina G/análise , Estudos de Coortes , Regulação para Baixo , Feminino , Fusobactérias/classificação , Bactérias Gram-Negativas/classificação , Humanos , Quinase I-kappa B/análise , Interleucina-6/análise , Masculino , Pessoa de Meia-Idade , Boca/imunologia , Mieloblastina/análise , Mieloblastina/antagonistas & inibidores , Proteína Adaptadora de Sinalização NOD2/análise , Doenças Periodontais/microbiologia , Peroxidase/análise , Proteobactérias/classificação , Fator de Necrose Tumoral alfa/análise
14.
J Cell Biol ; 204(2): 231-45, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24446482

RESUMO

Nuclear factor κB (NF-κB) essential modulator (NEMO), a regulatory component of the IκB kinase (IKK) complex, controls NF-κB activation through its interaction with ubiquitin chains. We show here that stimulation with interleukin-1 (IL-1) and TNF induces a rapid and transient recruitment of NEMO into punctate structures that are anchored at the cell periphery. These structures are enriched in activated IKK kinases and ubiquitinated NEMO molecules, which suggests that they serve as organizing centers for the activation of NF-κB. These NEMO-containing structures colocalize with activated TNF receptors but not with activated IL-1 receptors. We investigated the involvement of nondegradative ubiquitination in the formation of these structures, using cells deficient in K63 ubiquitin chains or linear ubiquitin chain assembly complex (LUBAC)-mediated linear ubiquitination. Our results indicate that, unlike TNF, IL-1 requires K63-linked and linear ubiquitin chains to recruit NEMO into higher-order complexes. Thus, different mechanisms are involved in the recruitment of NEMO into supramolecular complexes, which appear to be essential for NF-κB activation.


Assuntos
Quinase I-kappa B/metabolismo , Interleucina-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular Tumoral , Células HeLa , Humanos , Quinase I-kappa B/análise , Interleucina-1/análise , Interleucina-1/fisiologia , Quinases Associadas a Receptores de Interleucina-1/análise , Quinases Associadas a Receptores de Interleucina-1/metabolismo , NF-kappa B/análise , NF-kappa B/metabolismo , Receptores de Interleucina-1/análise , Receptores de Interleucina-1/metabolismo , Receptores do Fator de Necrose Tumoral/análise , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/fisiologia , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina/fisiologia , Ubiquitinação
15.
Inflammation ; 36(1): 242-50, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22968433

RESUMO

Stevioside, a diterpene glycoside component of Stevia rebaudiana, has been known to exhibit anti-inflammatory properties. To evaluate the effect and the possible mechanism of stevioside in lipopolysaccharide (LPS)-induced acute lung injury, male BALB/c mice were pretreated with stevioside or dexamethasone 1 h before intranasal instillation of LPS. Seven hours later, tumor necrosis factor-α, interleukin-1ß, and interleukin-6 in bronchoalveolar lavage fluid (BALF) were measured by using enzyme-linked immunosorbent assay. The number of total cells, neutrophils, and macrophages in the BALF were also determined. The right lung was excised for histological examination and analysis of myeloperoxidase activity and nitrate/nitrite content. Cyclooxygenase 2 (COX-2), inducible NO synthase (iNOS), nuclear factor-kappa B (NF-κB), inhibitory kappa B protein were detected by western blot. The results showed that stevioside markedly attenuated the LPS-induced histological alterations in the lung. Stevioside inhibited the production of pro-inflammatory cytokines and the expression of COX-2 and iNOS induced by LPS. In addition, not only was the wet-to-dry weight ratio of lung tissue significantly decreased, the number of total cells, neutrophils, and macrophages in the BALF were also significantly reduced after treatment with stevioside. Moreover, western blotting showed that stevioside inhibited the phosphorylation of IκB-α and NF-κB caused by LPS. Taken together, our results suggest that anti-inflammatory effect of stevioside against the LPS-induced acute lung injury may be due to its ability of inhibition of the NF-κB signaling pathway. Stevioside may be a promising potential therapeutic reagent for acute lung injury treatment.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Diterpenos do Tipo Caurano/uso terapêutico , Glucosídeos/uso terapêutico , Inflamação/tratamento farmacológico , NF-kappa B/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Contagem de Células , Ciclo-Oxigenase 2/análise , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Quinase I-kappa B/análise , Quinase I-kappa B/metabolismo , Inflamação/imunologia , Inflamação/patologia , Interleucina-1beta/análise , Interleucina-6/análise , Lipopolissacarídeos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/análise , Neutrófilos/imunologia , Nitratos/análise , Óxido Nítrico Sintase Tipo II/análise , Nitritos/análise , Peroxidase/análise , Fosforilação/efeitos dos fármacos , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/análise
16.
J Clin Endocrinol Metab ; 97(9): 3333-41, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22745245

RESUMO

CONTEXT: Sitagliptin is an inhibitor of the enzyme dipeptidyl peptidase-IV (DPP-IV), which degrades the incretins, glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide, and thus, sitagliptin increases their bioavailability. The stimulation of insulin and the suppression of glucagon secretion that follow exert a glucose lowering effect and hence its use as an antidiabetic drug. Because DPP-IV is expressed as CD26 on cell membranes and because CD26 mediates proinflammatory signals, we hypothesized that sitagliptin may exert an antiinflammatory effect. PATIENTS AND METHODS: Twenty-two patients with type 2 diabetes were randomized to receive either 100 mg daily of sitagliptin or placebo for 12 wk. Fasting blood samples were obtained at baseline and at 2, 4, and 6 hours after a single dose of sitagliptin and at 2, 4, 8, and 12 wk of treatment. RESULTS: Glycosylated hemoglobin fell significantly from 7.6 ± 0.4 to 6.9 ± 3% in patients treated with sitagliptin. Fasting glucagon-like peptide-1 concentrations increased significantly, whereas the mRNA expression in mononuclear cell of CD26, the proinflammatory cytokine, TNFα, the receptor for endotoxin, Toll-like receptor (TLR)-4, TLR-2, and proinflammatory kinases, c-Jun N-terminal kinase-1 and inhibitory-κB kinase (IKKß), and that of the chemokine receptor CCR-2 fell significantly after 12 wk of sitagliptin. TLR-2, IKKß, CCR-2, and CD26 expression and nuclear factor-κB binding also fell after a single dose of sitagliptin. There was a fall in protein expression of c-Jun N-terminal kinase-1, IKKß, and TLR-4 and in plasma concentrations of C-reactive protein, IL-6, and free fatty acids after 12 wk of sitagliptin. CONCLUSIONS: These effects are consistent with a potent and rapid antiinflammatory effect of sitagliptin and may potentially contribute to the inhibition of atherosclerosis. The suppression of CD26 expression suggests that sitagliptin may inhibit the synthesis of DPP-IV in addition to inhibiting its action.


Assuntos
Anti-Inflamatórios não Esteroides , Inibidores da Dipeptidil Peptidase IV/farmacologia , Pirazinas/farmacologia , Triazóis/farmacologia , Adulto , Idoso , Glicemia/análise , Glicemia/metabolismo , Western Blotting , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Separação Celular , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/fisiopatologia , Dipeptidil Peptidase 4/análise , Dipeptidil Peptidase 4/metabolismo , Método Duplo-Cego , Feminino , Peptídeo 1 Semelhante ao Glucagon/análise , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hemoglobinas Glicadas/análise , Humanos , Quinase I-kappa B/análise , Quinase I-kappa B/metabolismo , Interleucina-6/análise , Interleucina-6/metabolismo , MAP Quinase Quinase 4/análise , MAP Quinase Quinase 4/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Estudos Prospectivos , Receptores CCR2/análise , Receptores CCR2/metabolismo , Fosfato de Sitagliptina , Receptor 2 Toll-Like/análise , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/análise , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo
17.
Inflamm Res ; 61(5): 521-33, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22327510

RESUMO

OBJECTIVE: This study was designed to determine whether inhibition of heat shock protein 90 (HSP90) reduces pro-inflammatory mediator production by decreasing the nuclear factor (NF)-κB and Akt signaling pathways in immune-stimulated macrophages. METHODS: J774A.1 murine macrophages were treated with the HSP90 inhibitor 17-DMAG (0.01, 0.1 or 1 µM) prior to immune stimulation with lipopolysaccharide and interferon-γ. Expression of Akt, inhibitor of κB kinase (IKK), and heat shock proteins were measured in whole cell lysates by Western blotting. Phosphorylated Akt and inhibitor of κB (IκB) were measured in whole cell lysates by ELISA. Cell supernatants were analyzed for interleukin (IL)-6, tumor necrosis factor (TNF)-α and nitric oxide (NO). Translocation of NF-κB and heat shock factor (HSF)-1 was assessed by immunofluorescence. RESULTS: Treating cells with 17-DMAG reduced expression of Akt and IKK in immune-stimulated cells. 17-DMAG reduced nuclear translocation of NF-κB and reduced immune-stimulated production of IL-6, TNF-α and NO, but did not decrease inducible nitric oxide synthase expression. CONCLUSIONS: Our studies show that the immune-mediated NF-κB inflammatory cascade is blocked by the HSP90 inhibitor 17-DMAG. Due to the broad interaction of HSP90 with many pro-inflammatory kinase cascades, inhibition of HSP90 may provide a novel approach to reducing chronic inflammation.


Assuntos
Benzoquinonas/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lactamas Macrocíclicas/farmacologia , NF-kappa B/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Quinase I-kappa B/análise , Interferon gama/farmacologia , Interleucina-6/biossíntese , Lipopolissacarídeos/farmacologia , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/análise , Fator de Necrose Tumoral alfa/biossíntese
18.
Oncogene ; 30(2): 201-11, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20802527

RESUMO

Pancreatic adenocarcinoma upregulated factor (PAUF) is overproduced in certain types of cancer. However, little is known of the tumorigenic function of PAUF. In this study, we report the X-ray crystal structure of PAUF and reveal that PAUF is a mammalian lectin normally found in plant lectins. We also identify PAUF as an endogenous ligand of Toll-like receptor 2 (TLR2) and TLR4 by screening extracellular domain receptor pools. We further confirmed the specificity of the PAUF-TLR2 interaction. PAUF induces extracellular signal-regulated kinase (ERK) phosphorylation and activates the IKK-ß-mediated TPL2/MEK/ERK signaling pathway through TLR2. In agreement with the result of TLR2-mediated ERK activation by PAUF, PAUF induces increased expression of the protumorigenic cytokines RANTES and MIF in THP-1 cells. However, PAUF does not fully activate Iκ-B-α signaling pathways in THP-1 cells, and fails to translocate the p65 subunit of the nuclear factor-κB (NF-κB) complex into the nucleus, resulting in no NF-κB activation. Surprisingly, we found that PAUF also associated with the CXC chemokine receptor (CXCR4)-TLR2 complex and inhibited CXCR4-dependent, TLR2-mediated NF-κB activation. Together, these findings suggest that the new cancer-associated ligand, PAUF, may activate TLR-mediated ERK signaling to produce the protumorigenic cytokines, but inhibits TLR-mediated NF-κB signaling, thereby facilitating tumor growth and escape from innate immune surveillance.


Assuntos
Adenocarcinoma/secundário , Lectinas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores CXCR4/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células CHO , Quimiocina CCL5/análise , Quimiocina CCL5/metabolismo , Cricetinae , Cricetulus , Cristalografia , MAP Quinases Reguladas por Sinal Extracelular/análise , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/análise , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Oxirredutases Intramoleculares/análise , Oxirredutases Intramoleculares/metabolismo , Lectinas/química , MAP Quinase Quinase Quinases/análise , MAP Quinase Quinase Quinases/metabolismo , Fatores Inibidores da Migração de Macrófagos/análise , Fatores Inibidores da Migração de Macrófagos/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/metabolismo , Especificidade por Substrato , Regulação para Cima
19.
J Allergy Clin Immunol ; 126(1): 127-32.e7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20542322

RESUMO

BACKGROUND: Nuclear factor-kappaB (NF-kappaB) is a key transcription factor that regulates both innate and adaptive immunity as well as ectodermal development. Mutations in the coding region of the IkappaB kinase gamma/NF-kappaB essential modifier (NEMO) gene cause X-linked ectodermal dysplasia with immunodeficiency. OBJECTIVE: To determine the genetic cause of recurrent sinopulmonary infections and dysgammaglobulinemia in a patient with a normal NEMO coding sequence and his affected brother. METHODS: TNF-alpha and IFN-alpha production in response to Toll-like receptor (TLR) stimulation was analyzed by ELISA, NEMO mRNA levels were measured by quantitative PCR, and NEMO protein expression was measured by Western blotting. NF-kappaB activation was assessed by nuclear translocation of p65 and luciferase reporter gene assays. RESULTS: TLR-induced TNF-alpha and IFN-alpha production by PBMCs was impaired in the patient and his brother. Sequencing of the patient's NEMO gene revealed a novel mutation in the 5' untranslated region, which was also present in the brother, resulting in abnormally spliced transcripts and a 4-fold reduction in mRNA levels. NEMO protein levels in EBV transformed B cells and fibroblasts from the index patient were 8-fold lower than normal controls. NF-kappaB p65 nuclear translocation in the patient's EBV B cells after TLR7 ligation was defective. NF-kappaB-dependent luciferase gene expression in IL-1-stimulated fibroblasts from the patient was impaired. CONCLUSION: This is the first description of immune deficiency resulting from low expression of a normal NEMO protein.


Assuntos
Regiões 5' não Traduzidas/genética , Quinase I-kappa B/genética , Síndromes de Imunodeficiência/etiologia , Mutação , Criança , Citocinas/biossíntese , Humanos , Quinase I-kappa B/análise , Proteínas I-kappa B/metabolismo , Síndromes de Imunodeficiência/genética , Interleucina-1beta/farmacologia , Masculino , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fosforilação , Sítios de Splice de RNA , RNA Mensageiro/análise , Receptores Toll-Like/fisiologia
20.
J Dent Res ; 88(9): 835-40, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19767581

RESUMO

The inflammation observed in the dental pulp of teeth with deep caries lesions is characterized by a significant increase in blood vessel density. It is known that lipoteichoic acid (LTA) from Gram-positive cariogenic bacteria induces expression of vascular endothelial growth factor (VEGF) in dental pulp cells. The hypothesis underlying this study was that LTA induces VEGF expression in dental pulp cells through TLR2 and PI3k/Akt signaling. Odontoblast-like cells (MDPC-23) and undifferentiated pulp cells (OD-21) were exposed to LTA from Streptococcus sanguis, and the role of TLR2, PI3K/Akt, and IKK signaling in LTA-induced VEGF expression was evaluated. These studies demonstrated that TLR2 signaling through the PI3K-Akt pathway is necessary for LTA-induced VEGF expression in pulp cells. In contrast, inhibition of IKK signaling did not prevent VEGF up-regulation in response to LTA. Understanding signaling pathways triggered by cariogenic bacteria may reveal novel therapeutic targets for the clinical management of pulpitis.


Assuntos
Cárie Dentária/microbiologia , Polpa Dentária/microbiologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Streptococcus sanguis/fisiologia , Animais , Linhagem Celular , Polpa Dentária/irrigação sanguínea , Polpa Dentária/citologia , Fibroblastos/efeitos dos fármacos , Gengiva/citologia , Gengiva/efeitos dos fármacos , Quinase I-kappa B/análise , Quinase I-kappa B/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Microvasos/efeitos dos fármacos , Microvasos/patologia , Odontoblastos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/análise , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/análise , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Pulpite/microbiologia , Pulpite/patologia , Ácidos Teicoicos/farmacologia , Receptor 2 Toll-Like/análise , Receptor 2 Toll-Like/efeitos dos fármacos , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA