Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3966, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729935

RESUMO

Rhenium (Re) and uranium (U) are essential proxies in reconstructing past oceanic oxygenation evolution. However, their removal in continental shelf sediments, hotspots of early diagenesis, were previously treated as quantitatively unimportant sinks in the ocean. Here we examine the sedimentary reductive removal of Re and U and their coupling with organic carbon decomposition, utilizing the 224Ra/228Th disequilibria within the East China Sea shelf. We identified positive correlations between their removal fluxes and the rates of sediment oxygen consumption or organic carbon decomposition. These correlations enable an evaluation of global shelf reductive sinks that are comparable to (for Re) or higher than (~4-fold for U) previously established suboxic/anoxic sinks. These findings suggest potential imbalances in the modern budgets of Re and U, or perhaps a substantial underestimation of their sources. Our study thus highlights shelf sedimentary reductive removal as critical yet overlooked sinks for Re and U in the modern ocean.

2.
Chem Biol Drug Des ; 103(3): e14511, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38508859

RESUMO

Docetaxel (DTX) is a semi-synthetic analogue of paclitaxel which has attracted extensive attention in the treatment of cancer. However, the current clinically used DTX formulations display low tumor targeting ability, leading to unsatisfactory therapeutic outcomes with adverse effects, which poses significant challenges to the clinical application. In this study, three galactosamine (Gal) and docetaxel conjugates with different linkers were synthesized, namely DTX-(suc-Gal)2, DTX-(DTDPA-Gal)2, and DTX-(DSeDPA-Gal)2. These three conjugates were characterized by 1H NMR, FT-IR and HRMS. The in vitro drug release study shows that DTX-(DTDPA-Gal)2 and DTX-(DSeDPA-Gal)2 exhibit glutathione (GSH)-responsive drug release and DTX-(DSeDPA-Gal)2 displays higher GSH-responsiveness. The in vitro antitumor activity study shows that DTX-(DTDPA-Gal)2 and DTX-(DSeDPA-Gal)2 exhibit enhanced cytotoxicity, cell apoptosis rate and G2/M phase arrest against HepG2 cells as compared to DTX-(suc-Gal)2, DTX-(DSeDPA-Gal)2 displays the highest cytotoxicity, cell apoptosis rate and G2/M phase arrest among these three conjugates. In addition, DTX-(DSeDPA-Gal)2 exhibits higher selectivity to HepG2 cells as compared to free DTX. The DTX-(DSeDPA-Gal)2 developed in this study has been proven to be an effective DTX conjugate for selective killing hepatoma cells.


Assuntos
Antineoplásicos , Docetaxel/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Galactosamina , Espectroscopia de Infravermelho com Transformada de Fourier , Taxoides/farmacologia , Taxoides/química , Portadores de Fármacos/química , Linhagem Celular Tumoral
3.
Microbiol Mol Biol Rev ; 88(1): e0005222, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38451081

RESUMO

SUMMARYGroup A Streptococcus (GAS), also known as Streptococcus pyogenes, is a clinically well-adapted human pathogen that harbors rich virulence determinants contributing to a broad spectrum of diseases. GAS is capable of invading epithelial, endothelial, and professional phagocytic cells while evading host innate immune responses, including phagocytosis, selective autophagy, light chain 3-associated phagocytosis, and inflammation. However, without a more complete understanding of the different ways invasive GAS infections develop, it is difficult to appreciate how GAS survives and multiplies in host cells that have interactive immune networks. This review article attempts to provide an overview of the behaviors and mechanisms that allow pathogenic GAS to invade cells, along with the strategies that host cells practice to constrain GAS infection. We highlight the counteractions taken by GAS to apply virulence factors such as streptolysin O, nicotinamide-adenine dinucleotidase, and streptococcal pyrogenic exotoxin B as a hindrance to host innate immune responses.


Assuntos
Infecções Estreptocócicas , Streptococcus pyogenes , Humanos , Infecções Estreptocócicas/patologia , Imunidade Inata , Fatores de Virulência , Fagocitose
4.
Sensors (Basel) ; 24(2)2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-38257674

RESUMO

During the COVID-19 pandemic, the number of cases continued to rise. As a result, there was a growing demand for alternative control methods to traditional buttons or touch screens. However, most current gesture recognition technologies rely on machine vision methods. However, this method can lead to suboptimal recognition results, especially in situations where the camera is operating in low-light conditions or encounters complex backgrounds. This study introduces an innovative gesture recognition system for large movements that uses a combination of millimeter wave radar and a thermal imager, where the multi-color conversion algorithm is used to improve palm recognition on the thermal imager together with deep learning approaches to improve its accuracy. While the user performs gestures, the mmWave radar captures point cloud information, which is then analyzed through neural network model inference. It also integrates thermal imaging and palm recognition to effectively track and monitor hand movements on the screen. The results suggest that this combined method significantly improves accuracy, reaching a rate of over 80%.


Assuntos
COVID-19 , Gestos , Humanos , Pandemias , Algoritmos , COVID-19/diagnóstico , Mãos/diagnóstico por imagem
5.
Front Neurosci ; 16: 954332, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36051644

RESUMO

Background: Freezing of gait (FOG) in multiple system atrophy (MSA) is characterized by a higher risk of falls and a reduced quality of life; however, the mechanisms underlying these effects have yet to be identified by neuroimaging. The aim of this study was to investigate the differences in functional network when compared between MSA patients with and without freezing. Methods: Degree centrality (DC) based on the resting-state functional magnetic resonance imaging was computed in 65 patients with MSA and 36 healthy controls. Brain regions with statistically different DC values between groups were selected as seed points for a second seed-based functional connectivity (FC) analysis. The relationships between brain activity (DC and FC alterations) and the severity of freezing symptoms were then investigated in the two groups of patients with MSA. Results: Compared to MSA patients without FOG symptoms (MSA-nFOG), patients with MSA-FOG showed an increased DC in the left middle temporal gyrus but a reduced DC in the right superior pole temporal gyrus, left anterior cingulum cortex, left thalamus, and right middle frontal gyrus. Furthermore, in patients with MSA-FOG, the DC in the left thalamus was negatively correlated with FOG scores. Using the left thalamus as a seed, secondary seed-based functional connectivity analysis revealed that patients with MSA-FOG commonly showed the left thalamus-based FC abnormalities in regions related to cognition and emotion. In contrast to the patients with MSA-nFOG, patients with MSA-FOG showed an increased FC between the left thalamus and the left middle temporal gyrus (MTG), right inferior parietal lobule (IPL), bilateral cerebellum_8, and left precuneus. Conclusion: Freezing of gait is associated with centrality of the impaired thalamus network. Abnormal FC between the thalamus and left MTG, right IPL, bilateral cerebellum_8, and left precuneus was involved in FOG. These results provide new insight into the pathophysiological mechanism of FOG in MSA.

7.
Elife ; 92020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33185526

RESUMO

Legionella pneumophila causes a severe pneumonia known as Legionnaires' disease. During the infection, Legionella injects more than 300 effector proteins into host cells. Among them are enzymes involved in altering the host-ubiquitination system. Here, we identified two LegionellaOTU (ovarian tumor)-like deubiquitinases (LOT-DUBs; LotB [Lpg1621/Ceg23] and LotC [Lpg2529]). The crystal structure of the LotC catalytic core (LotC14-310) was determined at 2.4 Å. Unlike the classical OTU-family, the LOT-family shows an extended helical lobe between the Cys-loop and the variable loop, which defines them as a unique class of OTU-DUBs. LotB has an additional ubiquitin-binding site (S1'), which enables the specific cleavage of Lys63-linked polyubiquitin chains. By contrast, LotC only contains the S1 site and cleaves different species of ubiquitin chains. MS analysis of LotB and LotC identified different categories of host-interacting proteins and substrates. Together, our results provide new structural insights into bacterial OTU-DUBs and indicate distinct roles in host-pathogen interactions.


Assuntos
Bactérias/enzimologia , Enzimas Desubiquitinantes/metabolismo , Linhagem Celular , Enzimas Desubiquitinantes/genética , Escherichia coli , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Legionella , Legionelose , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Ubiquitinação
8.
Front Microbiol ; 11: 117, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117141

RESUMO

Group A streptococcus (GAS) is a versatile pathogen that causes a wide spectrum of diseases in humans. Invading host cells is a known strategy for GAS to avoid antibiotic killing and immune recognition. However, the underlying mechanisms of GAS resistance to intracellular killing need to be explored. Endothelial HMEC-1 cells were infected with GAS, methicillin-resistant Staphylococcus aureus (MRSA) and Salmonella Typhimurium under nicotinamide (NAM)-supplemented conditions. The intracellular NAD+ level and cell viability were respectively measured by NAD+ quantification kit and protease-based cytotoxicity assay. Moreover, the intracellular bacteria were analyzed by colony-forming assay, transmission electron microscopy, and confocal microscopy. We found that supplementation with exogenous nicotinamide during infection significantly inhibited the growth of intracellular GAS in endothelial cells. Moreover, the NAD+ content and NAD+/NADH ratio of GAS-infected endothelial cells were dramatically increased, whereas the cell cytotoxicity was decreased by exogenous nicotinamide treatment. After knockdown of the autophagy-related ATG9A, the intracellular bacterial load was increased in nicotinamide-treated endothelial cells. The results of Western blot and transmission electron microscopy also revealed that cells treated with nicotinamide can increase autophagy-associated LC3 conversion and double-membrane formation during GAS infection. Confocal microscopy images further showed that more GAS-containing vacuoles were colocalized with lysosome under nicotinamide-supplemented conditions than without nicotinamide treatment. In contrast to GAS, supplementation with exogenous nicotinamide did not effectively inhibit the growth of MRSA or S. Typhimurium in endothelial cells. These results indicate that intracellular NAD+ homeostasis is crucial for controlling intracellular GAS infection in endothelial cells. In addition, nicotinamide may be a potential new therapeutic agent to overcome persistent infections of GAS.

9.
mBio ; 10(5)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575768

RESUMO

Group A streptococcus (GAS) is an important human pathogen which can cause fatal diseases after invasion into the bloodstream. Although antibiotics and immune surveillance are the main defenses against GAS infection, GAS utilizes internalization into cells as a major immune evasion strategy. Our previous findings revealed that light chain 3 (LC3)-associated single membrane GAS-containing vacuoles in endothelial cells are compromised for bacterial clearance due to insufficient acidification after fusion with lysosomes. However, the characteristics and the activation mechanisms of these LC3-positive compartments are still largely unknown. In the present study, we demonstrated that the LC3-positive GAS is surrounded by single membrane and colocalizes with NADPH oxidase 2 (NOX2) complex but without ULK1, which are characteristics of LC3-associated phagocytosis (LAP). Inhibition of NOX2 or reactive oxygen species (ROS) significantly reduces GAS multiplication and enhances autolysosome acidification in endothelial cells through converting LAP to conventional xenophagy, which is revealed by enhancement of ULK1 recruitment, attenuation of p70s6k phosphorylation, and formation of the isolation membrane. We also clarify that the inactivation of mTORC1, which is the initiation signal of autophagy, is inhibited by NOX2- and ROS-activated phosphatidylinositol 3-kinase (PI3K)/AKT and MEK/extracellular signal-regulated kinase (ERK) pathways. In addition, streptolysin O (SLO) of GAS is identified as a crucial inducer of ROS for ß1 integrin-mediated LAP induction. After downregulation of ß1 integrin, GAS multiplication is reduced, accompanied with LAP inhibition and xenophagy induction. These results demonstrate that GAS infection preferentially induces ineffective LAP to evade xenophagic killing in endothelial cells through the SLO/ß1 integrin/NOX2/ROS pathway.IMPORTANCE Our previous reports showed that the LC3-associated GAS-containing single membrane vacuoles are inefficient for bacterial clearance in endothelial cells, which may result in bacteremia. However, the characteristics and the induction mechanisms of these LC3-positive vacuoles are still largely unknown. Here we provide the first evidence that these LC3-positive GAS-containing single membrane compartments appear to be LAPosomes, which are induced by NOX2 and ROS. Through NOX2- and ROS-mediated signaling, GAS preferentially induces LAP and inhibits bacteriostatic xenophagy in endothelial cells. We also provide the first demonstration that ß1 integrin acts as the receptor for LAP induction through GAS-produced SLO stimulation in endothelial cells. Our findings reveal the underlying mechanisms of LAP induction and autophagy evasion for GAS multiplication in endothelial cells.


Assuntos
Células Endoteliais/microbiologia , Macroautofagia , Streptococcus pyogenes/fisiologia , Estreptolisinas/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Humanos , Integrina beta1/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , NADPH Oxidase 2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Vacúolos/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-29581121

RESUMO

Group A Streptococcus (GAS) is an important human pathogen that causes a wide spectrum of diseases, including necrotizing fasciitis and streptococcal toxic shock syndrome. Dextromethorphan (DM), an antitussive drug, has been demonstrated to efficiently reduce inflammatory responses, thereby contributing to an increased survival rate of GAS-infected mice. However, the anti-inflammatory mechanisms underlying DM treatment in GAS infection remain unclear. DM is known to exert neuroprotective effects through an NADPH oxidase-dependent regulated process. In the present study, membrane translocation of NADPH oxidase subunit p47phox and subsequent reactive oxygen species (ROS) generation induced by GAS infection were significantly inhibited via DM treatment in RAW264.7 murine macrophage cells. Further determination of proinflammatory mediators revealed that DM effectively suppressed inducible nitric oxide synthase (iNOS) expression and NO, tumor necrosis factor alpha, and interleukin-6 generation in GAS-infected RAW264.7 cells as well as in air-pouch-infiltrating cells from GAS/DM-treated mice. GAS infection caused AKT dephosphorylation, glycogen synthase kinase-3ß (GSK-3ß) activation, and subsequent NF-κB nuclear translocation, which were also markedly inhibited by treatment with DM and an NADPH oxidase inhibitor, diphenylene iodonium. These results suggest that DM attenuates GAS infection-induced overactive inflammation by inhibiting NADPH oxidase-mediated ROS production that leads to downregulation of the GSK-3ß/NF-κB/NO signaling pathway.


Assuntos
Dextrometorfano/uso terapêutico , Óxido Nítrico Sintase Tipo II/metabolismo , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/enzimologia , Animais , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Oniocompostos/farmacologia , Oxirredução/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Infecções Estreptocócicas/metabolismo , Células THP-1
11.
Neuromolecular Med ; 20(1): 63-72, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29299869

RESUMO

Acute ischemic stroke causes a high rate of deaths and permanent neurological deficits in survivors. Current interventional treatment, in the form of enzymatic thrombolysis, benefits only a small percentage of patients. Brain ischemia triggers mobilization of innate immunity, specifically the complement system and Toll-like receptors (TLRs), ultimately leading to an exaggerated inflammatory response. Here we demonstrate that intravenous immunoglobulin (IVIG), a scavenger of potentially harmful complement fragments, and C1-esterase inhibitor (C1-INH), an inhibitor of complement activation, exert a beneficial effect on the outcome of experimental brain ischemia (I) and reperfusion (R) injury induced by transient occlusion of middle cerebral artery in mice. Both IVIG and C1-INH significantly and in a dose-responsive manner reduced brain infarction size, neurological deficit and mortality when administered to male mice 30 min before ischemia or up to 6 h after the onset of reperfusion. When combined, suboptimal doses of IVIG and C1-INH potentiated each other's neuroprotective therapeutic effects. Complement C3 and TLR2 signals were colocalized and significantly greater in brain cells adjacent to infracted brain lesions when compared to the corresponding regions of the contralateral hemisphere and to control (sham) mice. Treatment with IVIG and C1-INH effectively reduced deposition of C3b and downregulated excessive TLR2 and p-JNK1 expression at the site of I/R injury. Taken together, these results provide a rationale for potential use of IVIG and C1-INH, alone or in combination with ischemic stroke and other neurological conditions that involve inappropriately activated components of the innate immune system.


Assuntos
Proteína Inibidora do Complemento C1/uso terapêutico , Inativadores do Complemento/uso terapêutico , Transtornos Neurológicos da Marcha/prevenção & controle , Imunoglobulinas Intravenosas/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Ativação do Complemento/efeitos dos fármacos , Proteína Inibidora do Complemento C1/administração & dosagem , Complemento C3b/análise , Inativadores do Complemento/administração & dosagem , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Feminino , Transtornos Neurológicos da Marcha/etiologia , Imunoglobulinas Intravenosas/administração & dosagem , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Método Simples-Cego , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/genética , Regulação para Cima
12.
Mol Neurobiol ; 55(2): 1082-1096, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28092085

RESUMO

Multi-protein complexes, termed "inflammasomes," are known to contribute to neuronal cell death and brain injury following ischemic stroke. Ischemic stroke increases the expression and activation of nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) Pyrin domain containing 1 and 3 (NLRP1 and NLRP3) inflammasome proteins and both interleukin (IL)-1ß and IL-18 in neurons. In this study, we provide evidence that activation of either the NF-κB and MAPK signaling pathways was partly responsible for inducing the expression and activation of NLRP1 and NLRP3 inflammasome proteins and that these effects can be attenuated using pharmacological inhibitors of these two pathways in neurons and brain tissue under in vitro and in vivo ischemic conditions, respectively. Moreover, these findings provided supporting evidence that treatment with intravenous immunoglobulin (IVIg) preparation can reduce activation of the NF-κB and MAPK signaling pathways resulting in decreased expression and activation of NLRP1 and NLRP3 inflammasomes, as well as increasing expression of anti-apoptotic proteins, Bcl-2 and Bcl-xL, in primary cortical neurons and/or cerebral tissue under in vitro and in vivo ischemic conditions. In summary, these results provide compelling evidence that both the NF-κB and MAPK signaling pathways play a pivotal role in regulating the expression and activation of NLRP1 and NLRP3 inflammasomes in primary cortical neurons and brain tissue under ischemic conditions. In addition, treatment with IVIg preparation decreased the activation of the NF-κB and MAPK signaling pathways, and thus attenuated the expression and activation of NLRP1 and NLRP3 inflammasomes in primary cortical neurons under ischemic conditions. Hence, these findings suggest that therapeutic interventions that target inflammasome activation in neurons may provide new opportunities in the future treatment of ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inflamassomos/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antracenos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Butadienos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Imidazóis/farmacologia , Inflamassomos/efeitos dos fármacos , Camundongos , NF-kappa B/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neurônios/efeitos dos fármacos , Nitrilas/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulfonas/farmacologia
13.
mBio ; 8(4)2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28743815

RESUMO

Group A streptococcus (GAS) is an important human pathogen that causes a wide variety of cutaneous and systemic infections. Although originally thought to be an extracellular bacterium, numerous studies have demonstrated that GAS can trigger internalization into nonimmune cells to escape from immune surveillance or antibiotic-mediated killing. Epithelial cells possess a defense mechanism involving autophagy-mediated targeting and killing of GAS within lysosome-fused autophagosomes. In endothelial cells, in contrast, we previously showed that autophagy is not sufficient for GAS killing. In the present study, we showed higher galectin-3 (Gal-3) expression and lower Gal-8 expression in endothelial cells than in epithelial cells. The recruitment of Gal-3 to GAS is higher and the recruitment of Gal-8 to GAS is lower in endothelial cells than in epithelial cells. We further showed that Gal-3 promotes GAS replication and diminishes the recruitment of Gal-8 and ubiquitin, the latter of which is a critical protein for autophagy sequestration. After knockdown of Gal-3 in endothelial cells, the colocalization of Gal-8, parkin, and ubiquitin-decorated GAS is significantly increased, as is the interaction of Gal-8 and parkin, an E3 ligase. Furthermore, inhibition of Gal-8 in epithelial cells attenuates recruitment of parkin; both Gal-8 and parkin contribute to ubiquitin recruitment and GAS elimination. Animal studies confirmed that Gal-3-knockout mice develop less-severe skin damage and that GAS replication can be detected only in the air pouch and not in organs and endothelial cells. These results demonstrate that Gal-3 inhibits ubiquitin recruitment by blocking Gal-8 and parkin recruitment, resulting in GAS replication in endothelial cells.IMPORTANCE In epithelial cells, GAS can be efficiently killed within the lysosome-fused autophaosome compartment. However, we previously showed that, in spite of LC-3 recruitment, the autophagic machinery is not sufficient for GAS killing in endothelial cells. In this report, we provide the first evidence that Gal-3, highly expressed in endothelial cells, blocks the tagging of ubiquitin to GAS by inhibiting recruitment of Gal-8 and parkin, leading to an enhancement of GAS replication. We also provide the first demonstration that Gal-8 can interact with parkin, the critical E3 ligase, for resistance to intracellular bacteria by facilitating the decoration of bacteria with ubiquitin chains. Our findings reveal that differential levels of Gal-3 and Gal-8 expression and recruitment to GAS between epithelial cells and endothelial cells may contribute to the different outcomes of GAS elimination or survival and growth of GAS in these two types of cells.


Assuntos
Galectina 3/metabolismo , Galectinas/metabolismo , Streptococcus pyogenes/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Células A549 , Animais , Autofagia , Proteínas Sanguíneas , Células Endoteliais/microbiologia , Células Epiteliais/microbiologia , Galectina 3/deficiência , Galectina 3/genética , Galectinas/antagonistas & inibidores , Galectinas/deficiência , Galectinas/genética , Inativação Gênica , Humanos , Camundongos , Camundongos Knockout , Interferência de RNA , Pele/microbiologia , Pele/patologia , Streptococcus pyogenes/crescimento & desenvolvimento , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
14.
PLoS Pathog ; 13(7): e1006444, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28683091

RESUMO

Group A Streptococcus (GAS) is deleterious pathogenic bacteria whose interaction with blood vessels leads to life-threatening bacteremia. Although xenophagy, a special form of autophagy, eliminates invading GAS in epithelial cells, we found that GAS could survive and multiply in endothelial cells. Endothelial cells were competent in starvation-induced autophagy, but failed to form double-membrane structures surrounding GAS, an essential step in xenophagy. This deficiency stemmed from reduced recruitment of ubiquitin and several core autophagy proteins in endothelial cells, as demonstrated by the fact that it could be rescued by exogenous coating of GAS with ubiquitin. The defect was associated with reduced NO-mediated ubiquitin signaling. Therefore, we propose that the lack of efficient clearance of GAS in endothelial cells is caused by their intrinsic inability to target GAS with ubiquitin to promote autophagosome biogenesis for xenophagy.


Assuntos
Autofagia , Células Endoteliais/citologia , Infecções Estreptocócicas/fisiopatologia , Streptococcus pyogenes/fisiologia , Linhagem Celular , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Fagossomos/metabolismo , Fagossomos/microbiologia , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Ubiquitina/metabolismo
15.
Sci Rep ; 7(1): 5676, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28720835

RESUMO

Annexin A2 (ANXA2), a phospholipid-binding protein, has multiple biological functions depending on its cellular localization. We previously demonstrated that IFN-γ-triggered ANXA2 secretion is associated with exosomal release. Here, we show that IFN-γ-induced autophagy is essential for the extracellular secretion of ANXA2 in lung epithelial cells. We observed colocalization of ANXA2-containing autophagosomes with multivesicular bodies (MVBs) after IFN-γ stimulation, followed by exosomal release. IFN-γ-induced exophagic release of ANXA2 could not be observed in ATG5-silenced or mutant RAB11-expressing cells. Furthermore, knockdown of RAB8A and RAB27A, but not RAB27B, reduced IFN-γ-triggered ANXA2 secretion. Surface translocation of ANXA2 enhanced efferocytosis by epithelial cells, and inhibition of different exophagic steps, including autophagosome formation, fusion of autophagosomes with MVBs, and fusion of amphisomes with plasma membrane, reduced ANXA2-mediated efferocytosis. Our data reveal a novel route of IFN-γ-induced exophagy of ANXA2.


Assuntos
Anexina A2/metabolismo , Autofagia/fisiologia , Interferon gama/farmacologia , Pulmão/efeitos dos fármacos , Células A549 , Células Epiteliais/efeitos dos fármacos , Células HEK293 , Humanos , Células Jurkat , Corpos Multivesiculares , Fagocitose , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP/metabolismo
16.
Sci Rep ; 7(1): 91, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273893

RESUMO

Activated microglial cells are present in dengue virus (DENV)-infected brains; however, the possible effects of DENV on microglia remain unclear. Here, we demonstrated DENV caused infection, including viral entry, RNA replication, viral protein expression, and virus release, in the murine microglial cell line BV2. DENV infection caused an increase in the formation of the multipolar phenotype in vitro and in vivo without affecting cell growth and cytotoxicity. DENV infection considerably increased cell motility and disrupting either actin filaments or clathrin retarded such effect. Increase in cell migration was only occurred by DENV infection following a clathrin-regulated endocytosis of DENV entry. Ultraviolet-inactivated DENV did not affect cell migration, and pharmacologically blocking toll-like receptor (TLR) 3 and TLR3-related signaling pathways reduced the DENV-induced increase in cell migration. These results demonstrate an advanced effect of DENV infection on microglial migration via a mechanism involving viral entry, RNA release, and TLR3 signal activation.


Assuntos
Vírus da Dengue/patogenicidade , Dengue/virologia , Microglia/citologia , RNA Viral/genética , Receptor 3 Toll-Like/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Linhagem Celular , Movimento Celular , Clatrina/metabolismo , Dengue/metabolismo , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Endocitose , Humanos , Camundongos , Microglia/metabolismo , Microglia/virologia , Modelos Biológicos , Transdução de Sinais , Internalização do Vírus , Replicação Viral
17.
Sci Rep ; 7: 42998, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28216632

RESUMO

Dengue is one of the most significant mosquito-borne virus diseases worldwide, particularly in tropical and subtropical regions. This study sought to examine the antiviral activity of resveratrol (RESV), a phytoalexin secreted naturally by plants, against dengue virus (DENV) infection. Our data showed that RESV inhibits the translocation of high mobility group box 1 (HMGB1), a DNA binding protein that normally resides in the nucleus, into the cytoplasm and extracellular milieu. HMGB1 migrates out of the nucleus during DENV infection. This migration is inhibited by RESV treatment and is mediated by induction of Sirt1 which leads to the retention of HMGB1 in the nucleus and consequently helps in the increased production of interferon-stimulated genes (ISGs). Nuclear HMGB1 was found to bind to the promoter region of the ISG and positively regulated the expression of ISG. The enhanced transcription of ISGs by nuclear HMGB1 thus contributes to the antiviral activity of RESV against DENV. To the best of our knowledge, this is the first report to demonstrate that RESV antagonizes DENV replication and that nuclear HMGB1 plays a role in regulating ISG production.


Assuntos
Antivirais/farmacologia , Proteína HMGB1/metabolismo , Interferon Tipo I/metabolismo , Estilbenos/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/uso terapêutico , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Dengue/tratamento farmacológico , Dengue/patologia , Dengue/virologia , Vírus da Dengue/isolamento & purificação , Vírus da Dengue/fisiologia , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Humanos , Interferon Tipo I/genética , Interferon beta/genética , Interferon beta/metabolismo , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Resveratrol , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Sirtuína 1/metabolismo , Estilbenos/uso terapêutico
18.
ACS Appl Mater Interfaces ; 8(38): 25069-77, 2016 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-27589088

RESUMO

One of the key challenges in engineering neural tissues for cell-based therapies is to develop a biocompatible scaffold material to direct neural stem cell (NSC) behaviors. One great advantage for a scaffold would be to induce NSC migration toward pathological sites during regeneration and repair. In particular, the inflammatory responses in the pathological zone, which are mainly mediated by microglia in the central nervous system, affect the repair capacity of NSCs through NSC migration. Recently, graphene was used as a neural interface and scaffold material, but few studies have addressed the relationship between microglia and NSCs in a graphene culture system. In this study, we used a combination of immunofluorescence, Western blotting, enzyme-linked immunosorbent assays, and scanning electron microscopy to investigate how conditioned medium (CM) produced from microglia grown on two-dimensional graphene (2D-G) films or three-dimensional graphene (3D-G) foams govern NSC migration. The results revealed that the CM produced by microglia grown in 3D-G cultures could promote neurosphere formation, facilitate NSC migration from the neurospheres, and increase single cell polarization by activating the stromal cell-derived factor 1 α (SDF-1α)/CXC chemokine receptor 4 (CXCR4) signaling pathway and enhancing cell adhesion on the substrate. By contrast, the 2D-G CM failed to achieve these results. Our study suggests the great potential of 3D-G as a neural scaffold for NSC-based therapy in tissue engineering and regenerative medicine.


Assuntos
Células-Tronco Neurais , Movimento Celular , Quimiocina CXCL12 , Grafite , Microglia , Receptores CXCR4
19.
Sci Rep ; 6: 32000, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27561946

RESUMO

Infection by the dengue virus (DENV) threatens global public health due to its high prevalence and the lack of effective treatments. Host factors may contribute to the pathogenesis of DENV; herein, we investigated the role of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is activated by DENV in mononuclear phagocytes. DENV infection selectively activates Nrf2 following nuclear translocation. Following endoplasmic reticular (ER) stress, protein kinase R-like ER kinase (PERK) facilitated Nrf2-mediated transcriptional activation of C-type lectin domain family 5, member A (CLEC5A) to increase CLEC5A expression. Signaling downstream of the Nrf2-CLEC5A interaction enhances Toll-like receptor 3 (TLR3)-independent tumor necrosis factor (TNF)-α production following DENV infection. Forced expression of the NS2B3 viral protein induces Nrf2 nuclear translocation/activation and CLEC5A expression which increases DENV-induced TNF-α production. Animal studies confirmed Nrf2-induced CLEC5A and TNF-α in brains of DENV-infected mice. These results demonstrate that DENV infection causes Nrf2-regulated TNF-α production by increasing levels of CLEC5A.


Assuntos
Vírus da Dengue/fisiologia , Lectinas Tipo C/metabolismo , Monócitos/virologia , Fator 2 Relacionado a NF-E2/fisiologia , Receptores de Superfície Celular/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Cricetinae , Humanos , Camundongos , Monócitos/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Ativação Transcricional , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
20.
Sci Rep ; 6: 27670, 2016 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-27279150

RESUMO

Patients with dengue virus (DENV) infection may also present acute viral encephalitis through an unknown mechanism. Here, we report that encephalitic DENV-infected mice exhibited progressive hunchback posture, limbic seizures, limbic weakness, paralysis, and lethality 7 days post-infection. These symptoms were accompanied by CNS inflammation, neurotoxicity, and blood-brain barrier destruction. Microglial cells surrounding the blood vessels and injured hippocampus regions were activated by DENV infection. Pharmacologically depleting microglia unexpectedly increased viral replication, neuropathy, and mortality in DENV-infected mice. In microglia-depleted mice, the DENV infection-mediated expression of antiviral cytokines and the infiltration of CD8-positive cytotoxic T lymphocytes (CTLs) was abolished. DENV infection prompted the antigen-presenting cell-like differentiation of microglia, which in turn stimulated CTL proliferation and activation. These results suggest that microglial cells play a key role in facilitating antiviral immune responses against DENV infection and acute viral encephalitis.


Assuntos
Dengue/complicações , Encefalite Viral/imunologia , Microglia/imunologia , Aedes , Animais , Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Cultivadas , Cricetinae , Citocinas/genética , Citocinas/metabolismo , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , Encefalite Viral/etiologia , Hipocampo/citologia , Hipocampo/virologia , Camundongos , Camundongos Endogâmicos ICR , Microglia/virologia , Linfócitos T Citotóxicos/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...