Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Negl Trop Dis ; 16(9): e0010743, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36070318

RESUMO

Mosquito saliva proteins modulate the human immune and hemostatic systems and control mosquito-borne pathogenic infections. One mechanism through which mosquito proteins may influence host immunity and hemostasis is their interactions with key human receptor proteins that may act as receptors for or coordinate attacks against invading pathogens. Here, using pull-down assays and proteomics-based mass spectrometry, we identified 11 Ae. aegypti salivary gland proteins (SGPs) (e.g., apyrase, Ae. aegypti venom allergen-1 [AaVA-1], neutrophil stimulating protein 1 [NeSt1], and D7 proteins), that interact with one or more of five human receptor proteins (cluster of differentiation 4 [CD4], CD14, CD86, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin [DC-SIGN], and Toll-like receptor 4 [TLR4]). We focused on CD4- and DC-SIGN-interacting proteins and confirmed that CD4 directly interacts with AaVA-1, D7, and NeST1 recombinant proteins and that AaVA-1 showed a moderate interaction with DC-SIGN using ELISA. Bacteria responsive protein 1 (AgBR1), an Ae. aegypti saliva protein reported to enhance ZIKV infection in humans but that was not identified in our pull-down assay moderately interacts with CD4 in the ELISA assay. Functionally, we showed that AaVA-1 and NeST1 proteins promoted activation of CD4+ T cells. We propose the possible impact of these interactions and effects on mosquito-borne viral infections such as dengue, Zika, and chikungunya viruses. Overall, this study provides key insight into the vector-host (protein-protein) interaction network and suggests roles for these interactions in mosquito-borne viral infections.


Assuntos
Aedes , Proteínas e Peptídeos Salivares , Alérgenos , Animais , Apirase , Humanos , Molécula 3 de Adesão Intercelular/metabolismo , Mosquitos Vetores , Proteínas Recombinantes/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Receptor 4 Toll-Like/metabolismo
2.
Br J Cancer ; 123(5): 730-741, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32528119

RESUMO

BACKGROUND: The anticancer potential of ibuprofen has created a broad interest to explore the clinical benefits of ibuprofen in cancer therapy. However, the current understanding of the molecular mechanisms involved in the anticancer potential of ibuprofen remains limited. METHODS: Cancer stemness assays to validate ibuprofen function in vitro and in vivo. Histone modification assays to check the effect of ibuprofen on histone acetylation/methylation, as well as the activity of HDAC and KDM6A/B. Inhibitors' in vivo assays to evaluate therapeutic effects of various inhibitors' combination manners. RESULTS: In our in vitro studies, we report that ibuprofen diminishes cancer cell stemness properties that include reducing the ALDH + subpopulation, side population and sphere formation in three cancer types. In our in vivo studies, we report that ibuprofen decreases tumour growth, metastasis and prolongs survival. In addition, our results showed that ibuprofen inhibits inflammation-related stemness gene expression (especially ICAM3) identified by a high-throughput siRNA platform. In regard to the underlying molecular mechanism of action, we report that ibuprofen reduces HDACs and histone demethylase (KDM6A/B) expression that mediates histone acetylation and methylation, and suppresses gene expression via a COX2-dependent way. In regard to therapeutic strategies, we report that ibuprofen combined HDAC/HDM inhibitors prevents cancer progression in vivo. CONCLUSIONS: The aforementioned findings suggest a molecular model that explains how ibuprofen diminishes cancer cell stemness properties. These may provide novel targets for therapeutic strategies involving ibuprofen in the prevention of cancer progression.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Histonas/metabolismo , Ibuprofeno/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células A549 , Acetilação/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Células Hep G2 , Histona Desacetilases/metabolismo , Humanos , Molécula 3 de Adesão Intercelular/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Distribuição Aleatória
3.
Molecules ; 24(9)2019 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-31083610

RESUMO

Optimal targeting of nanoparticles (NP) to dendritic cells (DCs) receptors to deliver cancer-specific antigens is key to the efficient induction of anti-tumour immune responses. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing tètanus toxoid and gp100 melanoma-associated antigen, toll-like receptor adjuvants were targeted to the DC-SIGN receptor in DCs by specific humanized antibodies or by ICAM3-Fc fusion proteins, which acts as the natural ligand. Despite higher binding and uptake efficacy of anti-DC-SIGN antibody-targeted NP vaccines than ICAM3-Fc ligand, no difference were observed in DC activation markers CD80, CD83, CD86 and CCR7 induced. DCs loaded with NP coated with ICAM3-Fc appeared more potent in activating T cells via cross-presentation than antibody-coated NP vaccines. This fact could be very crucial in the design of new cancer vaccines.


Assuntos
Vacinas Anticâncer/metabolismo , Células Dendríticas/metabolismo , Molécula 3 de Adesão Intercelular/metabolismo , Nanopartículas/química , Vacinas Anticâncer/química , Células Cultivadas , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Leucócitos/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptores de IgG/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
J Immunol ; 203(1): 148-157, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31118225

RESUMO

MicroRNAs are small, noncoding RNAs that function as posttranscriptional modulators of gene expression by binding target mRNAs and inhibiting translation. They are therefore crucial regulators of several biological as well as immunological events. Recently, miR-511-3p has been implicated in the development and differentiation of APCs, such as dendritic cells (DCs), and regulating several human diseases. Interestingly, miR-511-3p is embedded within the human MRC1 gene that encodes the mannose receptor. In this study, we sought to examine the impact of miR-511-3p up- or downregulation on human DC surface phenotype, cytokine profile, immunogenicity (using IDO activity as a surrogate), and downstream T cell polarization. Using gene silencing and a selection of microRNA mimics, we could successfully suppress or induce the expression of miR-511-3p in DCs. Consequently, we show for the first time, to our knowledge, that inhibition and/or overexpression of miR-511-3p has opposing effects on the expression levels of two key C-type lectin receptors, namely the mannose receptor and DC-specific ICAM 3 nonintegrin at protein and mRNA levels, thereby affecting C-type lectin receptor-induced modulation of IDO activity in DCs. Furthermore, we show that downregulation of miR-511-3p drives an anti-inflammatory DC response characterized by IL-10 production. Interestingly, the miR-511-3plow DCs also promoted IL-4 secretion and suppressed IL-17 in cocultures with autologous T cells. Together, our data highlight the potential role of miR-511 in regulating DC function and downstream events leading to Th polarization and immune modulation.


Assuntos
Células Dendríticas/fisiologia , Lectinas Tipo C/metabolismo , MicroRNAs/genética , Linfócitos T Auxiliares-Indutores/imunologia , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica , Humanos , Imunomodulação , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Molécula 3 de Adesão Intercelular/genética , Molécula 3 de Adesão Intercelular/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Ativação Linfocitária , RNA Interferente Pequeno/genética , Receptor Cross-Talk
5.
Int J Mol Sci ; 20(7)2019 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-30935090

RESUMO

BACKGROUND: In pregnancy, excessive inflammation and break down of immunologic tolerance can contribute to miscarriage. Endothelial cells (ECs) are able to orchestrate the inflammatory processes by secreting pro-inflammatory mediators and bactericidal factors by modulating leakiness and leukocyte trafficking, via the expression of adhesion molecules and chemokines. The aim of this study was to analyse the differences in the phenotype between microvascular ECs isolated from decidua (DECs) and ECs isolated from human skin (ADMECs). METHODS: DECs and ADMECs were characterized for their basal expression of angiogenic factors and adhesion molecules. A range of immunological responses was evaluated, such as vessel leakage, reactive oxygen species (ROS) production in response to TNF-α stimulation, adhesion molecules expression and leukocyte migration in response to TNF-α and IFN-γ stimulation. RESULTS: DECs produced higher levels of HGF, VEGF-A and IGFBP3 compared to ADMECs. DECs expressed adhesion molecules, ICAM-2 and ICAM-3, and a mild response to TNF-α was observed. Finally, DECs produced high levels of CXCL9/MIG and CXCL10/IP-10 in response to IFN-γ and selectively recruited Treg lymphocytes. CONCLUSION: DEC phenotype differs considerably from that of ADMECs, suggesting that DECs may play an active role in the control of immune response and angiogenesis at the foetal-maternal interface.


Assuntos
Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Pele/imunologia , Pele/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Decídua , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Imunofluorescência , Humanos , Técnicas In Vitro , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Molécula 3 de Adesão Intercelular/genética , Molécula 3 de Adesão Intercelular/metabolismo , Interferon gama/farmacologia , Neovascularização Patológica/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
6.
Pathol Oncol Res ; 24(1): 67-74, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28299711

RESUMO

Thymectomy is routinely carried out in patients with myasthenia gravis (MG) and thymomas. However, there is still a dispute as to whether MG patients with thymic hyperplasia should undergo thymectomy. We aimed to investigate the pathological findings in the thymus in patients with co-existing MG and thymic hyperplasia or thymomas treated with thymectomy, as well as effects of immunosuppression. Thirty-three patients with MG were selected and grouped accordingly: patients with no thymic abnormalities, patients with thymic hyperplasia, and patients with thymomas. All patients were treated with methylprednisolone alongside immunosuppression. A separate cohort of 24 MG patients with thymic hyperplasia or thymomas and treated with thymectomy were selected. As controls, 5 patients with thymomas or thymic carcinoma without MG were selected. Expression of CD5, extracellular regulated protein kinases1/2 mitogen activated protein kinase (ERK1/2MAPKs) and CD95 ligand (FasL) in the thymus was examined. Methylprednisolone and immunosuppressive therapy are highly effective in MG patients with normal thymus tissue and MG patients with thymic hyperplasia compared to MG patients with thymomas alone. CD5 expression was highest in MG patients with thymic hyperplasia, correlating with expression of ERK1/2MAPKs. FasL expression was similar across all groups. Thymomas may be distinguished from thymic hyperplasia by expression of CD5 and ERK1/2MAPKs. Thymectomy is the preferred treatment for MG patients with thymomas but may not be necessary in MG patients with thymic hyperplasia who are treated with immunosuppressive therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Imunossupressores/uso terapêutico , Miastenia Gravis/patologia , Timoma/patologia , Hiperplasia do Timo/patologia , Neoplasias do Timo/patologia , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Criança , Proteína Ligante Fas/metabolismo , Feminino , Seguimentos , Humanos , Molécula 3 de Adesão Intercelular/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miastenia Gravis/tratamento farmacológico , Miastenia Gravis/metabolismo , Prognóstico , Estudos Retrospectivos , Timoma/tratamento farmacológico , Timoma/metabolismo , Hiperplasia do Timo/tratamento farmacológico , Hiperplasia do Timo/metabolismo , Neoplasias do Timo/tratamento farmacológico , Neoplasias do Timo/metabolismo , Adulto Jovem
7.
EMBO Mol Med ; 9(11): 1482-1490, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28899929

RESUMO

Several lines of evidence indirectly suggest that antigenic stimulation through the B-cell receptor (BCR) supports chronic lymphocytic leukemia (CLL) development. In addition to self-antigens, a number of microbial antigens have been proposed to contribute to the selection of the immunoglobulins expressed in CLL. How pathogen-specific BCRs drive CLL development remains, however, largely unexplored. Here, we utilized mouse models of CLL pathogenesis to equip B cells with virus-specific BCRs and study the effect of antigen recognition on leukemia growth. Our results show that BCR engagement is absolutely required for CLL development. Unexpectedly, however, neither acute nor chronic exposure to virus-derived antigens influenced leukemia progression. Rather, CLL clones preferentially selected light chains that, when paired with virus-specific heavy chains, conferred B cells the ability to recognize a broad range of autoantigens. Taken together, our results suggest that pathogens may drive CLL pathogenesis by selecting and expanding pathogen-specific B cells that cross-react with one or more self-antigens.


Assuntos
Autoantígenos/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Cadeias Leves de Imunoglobulina/metabolismo , Imunoglobulinas/metabolismo , Molécula 3 de Adesão Intercelular/metabolismo , Leucemia Linfocítica Crônica de Células B/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise Serial de Proteínas , Proteínas Proto-Oncogênicas/genética , Baço/citologia , Baço/metabolismo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA