Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Math Biosci Eng ; 16(6): 7155-7176, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31698608

RESUMO

Modal identification involves primarily the determination of natural frequencies, damping ratios, mode shapes of a dynamic system, etc. It is usually regarded as an essential task in a wide branch of structural dynamics and civil engineering, such as structural vibration control and damage identification of buildings or bridges. There are many modal identification techniques. Basically, these techniques can be categorized into two groups: deterministic methods and Bayesian approaches. The first group can be used to provide deterministic (or optimal) estimations of modal parameters, but they are unable to quantify the estimation uncertainties. The second group is based on a usage of the Bayesian framework. Compared to the first group, the second group of methods has a typical merit of being able to offer uncertainty information of identified parameters, which is of great interests, or even necessary, for some follow-up studies. In this paper, both a deterministic method, i.e., a combination of spectral analysis, filtering and Random Decrement Technique (RDT), and a Bayesian method, i.e., Bayesian Spectral Density Approach (BSDA), are exploited to experimentally identify the modal parameters of a 303 m high-rise building that was subjected to a landfall typhoon. The validity and efficiency of each method is verified by comparing the two kinds of results. Meanwhile, the identified modal parameters are used for the serviceability assessment of this high-rise building against some frequency-specific criteria.

3.
J Neuroinflammation ; 16(1): 101, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092253

RESUMO

BACKGROUND: Blood-brain barrier (BBB) disruption and neuroinflammation are considered key mechanisms of pathogenic Escherichia coli invasion of the brain. However, the specific molecules involved in meningitic E. coli-induced BBB breakdown and neuroinflammatory response remain unclear. Our previous RNA-sequencing data from human brain microvascular endothelial cells (hBMECs) revealed two important host factors: platelet-derived growth factor-B (PDGF-B) and intercellular adhesion molecule-1 (ICAM-1), which were significantly upregulated in hBMECs after meningitic E. coli infection. Whether and how PDGF-B and ICAM-1 contribute to the development of E. coli meningitis are still unclear. METHODS: The western blot, real-time PCR, enzyme-linked immunosorbent assay, immunohistochemistry, and immunofluorescence were applied to verify the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in vivo and in vitro. Evan's blue assay and electric cell-substrate impedance sensing assay were combined to identify the effects of PDGF-B on BBB permeability. The CRISPR/Cas9 technology, cell-cell adhesion assay, and electrochemiluminescence assay were used to investigate the role of ICAM-1 in neuroinflammation subversion. RESULTS: We verified the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in mouse as well as monolayer hBMECs models. Functionally, we showed that the increase of PDGF-B may directly enhance the BBB permeability by decreasing the expression of tight junction proteins, and the upregulation of ICAM-1 contributed to neutrophils or monocytes recruitment as well as neuroinflammation subversion in response to meningitic E. coli infection. CONCLUSIONS: Our findings demonstrated the roles of PDGF-B and ICAM-1 in mediating bacterial-induced BBB damage as well as neuroinflammation, providing new concepts and potential targets for future prevention and treatment of bacterial meningitis.


Assuntos
Barreira Hematoencefálica/metabolismo , Infecções por Escherichia coli/metabolismo , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Linfocinas/biossíntese , Meningites Bacterianas/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Animais , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/patologia , Células Cultivadas , Escherichia coli , Infecções por Escherichia coli/patologia , Feminino , Meningites Bacterianas/patologia , Camundongos , Junções Íntimas/metabolismo , Junções Íntimas/microbiologia , Regulação para Cima/fisiologia
4.
J Neuroinflammation ; 15(1): 291, 2018 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-30340642

RESUMO

BACKGROUND: Bacterial meningitis remains a big threat to the integrity of the central nervous system (CNS), despite the advancements in antimicrobial reagents. Escherichia coli is a bacterial pathogen that can disrupt the CNS function, especially in neonates. E. coli meningitis occurs after bacteria invade the brain microvascular endothelial cells (BMECs) that form a direct and essential barrier restricting the entry of circulating microbes and toxins to the brain. Previous studies have reported on several cellular proteins that function during meningitic E. coli infections; however, more comprehensive investigations to elucidate the potential targets involved in E. coli meningitis are essential to better understand this disease and discover new treatments for it. METHODS: The isobaric tags for relative and absolute quantification (iTRAQ) approach coupled with LC-MS/MS were applied to compare and characterize the different proteomic profiles of BMECs in response to meningitic or non-meningitic E. coli strains. KEGG and gene ontology annotations, ingenuity pathways analysis, and functional experiments were combined to identify the key host molecules involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammatory responses. RESULTS: A total of 13 cellular proteins were found to be differentially expressed by meningitic E. coli strains PCN033 and RS218, including one that was also affected by HB101, a non-meningitic E. coli strain. Through bioinformatics analysis, we identified the macrophage migration inhibitory factor (MIF), granzyme A, NF-κB signaling, and mitogen-activated protein kinase (MAPK) pathways as being biologically involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammation. Functionally, we showed that MIF facilitated meningitic E. coli-induced production of cytokines and chemokines and also helped to disrupt the blood-brain barrier by decreasing the expression of tight junction proteins like ZO-1, occludin. Moreover, we demonstrated the significant activation of NF-κB and MAPK signaling in BMECs in response to meningitic E. coli strains, which dominantly determined the generation of the proinflammatory cytokines including IL-6, IL-8, TNF-α, and IL-1ß. CONCLUSIONS: Our work identified 12 host cellular targets that are affected by meningitic E. coli strains and revealed MIF to be an important contributor to meningitic E. coli-induced cytokine production and tight junction disruption, and also the NF-κB and MAPK signaling pathways that are mainly involved in the infection-induced cytokines production. Characterization of these distinct proteins and pathways in BMECs will facilitate further elucidation of meningitis-causing mechanisms in humans and animals, thereby enabling the development of novel preventative and therapeutic strategies against infection with meningitic E. coli.


Assuntos
Encéfalo/citologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteômica/métodos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Cultivadas , Biologia Computacional , Citocinas/genética , Citocinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/farmacologia , Fatores Inibidores da Migração de Macrófagos/química , Fatores Inibidores da Migração de Macrófagos/farmacologia , Meningite devida a Escherichia coli/metabolismo , Meningite devida a Escherichia coli/patologia , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia
5.
J Neuroinflammation ; 13(1): 274, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27756321

RESUMO

BACKGROUND: Streptococcus suis serotype 2 (SS2) is an important zoonotic bacterial pathogen in both humans and animals, which can cause high morbidity and mortality. Meningitis is one of the major clinical manifestations of SS2 infection. However, the specific process of SS2 meningitis and its molecular mechanisms remain unclear. Epidermal growth factor receptor (EGFR) has been reported to initiate transduction of intracellular signals and regulate host inflammatory responses. Whether and how EGFR contributes to the development of S. suis meningitis are currently unknown. METHODS: The tyrosine phosphorylation of cellular proteins, the transactivation of EGFR, as well as its dimerization, and the associated signal transduction pathways were investigated by immunoprecipitation and western blotting. Real-time quantitative PCR was used to investigate the transcriptional level of the ErbB family members, EGFR-related ligands, cytokines, and chemokines. The secretion of cytokines and chemokines in the serum and brain were detected by Q-Plex™ Chemiluminescent ELISA. RESULTS: We found an important role of EGFR in SS2 strain SC19-induced meningitis. SC19 increasingly adhered to human brain microvascular endothelial cells (hBMEC) and caused inflammatory lesions in the brain tissues, with significant induction and secretion of proinflammatory cytokines and chemokines in the serum and brains. SC19 infection of hBMEC induced tyrosine phosphorylation of cellular EGFR in a ligand-dependent manner involving the EGF-like ligand HB-EGF, amphiregulin (AREG), and epiregulin (EREG) and led to heterodimerization of EGFR/ErbB3. The EGFR transactivation did not participate in SS2 strain SC19 adhesion of hBMEC, as well as in bacterial colonization in vivo. However, its transactivation contributed to the bacterial-induced neuroinflammation, via triggering the MAPK-ERK1/2 and NF-κB signaling pathways in hBMEC that promote the production of proinflammatory cytokines and chemokines. CONCLUSIONS: We investigated for the first time the tyrosine phosphorylation of cellular proteins in response to SS2 strain SC19 infection of hBMEC and demonstrated the contribution of EGFR to SS2-induced neuroinflammation. These observations propose a novel mechanism involving EGFR in SS2-mediated inflammatory responses in the brain, and therefore, EGFR might be an important host target for further investigation and prevention of neuroinflammation caused by SS2 strains.


Assuntos
Encéfalo/metabolismo , Receptores ErbB/metabolismo , Meningite , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/fisiopatologia , Streptococcus suis/fisiologia , Ativação Transcricional/fisiologia , Anfirregulina/metabolismo , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Feminino , Humanos , Meningite/etiologia , Meningite/microbiologia , Meningite/fisiopatologia , Camundongos , Fosforilação/efeitos dos fármacos , Quinazolinas/farmacologia , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Infecções Estreptocócicas/microbiologia , Suínos , Tirosina/metabolismo , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...