Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Front Cell Infect Microbiol ; 12: 868407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35433510

RESUMEN

Dengue virus (DENV) causes dengue fever, which is prevalent in the tropical and subtropical regions, and in recent years, has resulted in several major epidemics. Vimentin, a cytoskeletal component involved in DENV infection, is significantly reorganized during infection. However, the mechanism underlying the association between DENV infection and vimentin is still poorly understood. We generated vimentin-knockout (Vim-KO) human brain microvascular endothelial cells (HBMECs) and a Vim-KO SV129 suckling mouse model, combining the dynamic vimentin changes observed in vitro and differences in disease course in vivo, to clarify the role of vimentin in DENV-2 infection. We found that the phosphorylation and solubility of vimentin changed dynamically during DENV-2 infection of HBMECs, suggesting the regulation of vimentin by DENV-2 infection. The similar trends observed in the phosphorylation and solubility of vimentin showed that these characteristics are related. Compared with that in control cells, the DENV-2 viral load was significantly increased in Vim-KO HBMECs, and after DENV-2 infection, Vim-KO SV129 mice displayed more severe disease signs than wild-type SV129 mice, as well as higher viral loads in their serum and brain tissue, demonstrating that vimentin can inhibit DENV-2 infection. Moreover, Vim-KO SV129 mice had more disordered cerebral cortical nerve cells, confirming that Vim-KO mice were more susceptible to DENV-2 infection, which causes severe brain damage. The findings of our study help clarify the mechanism by which vimentin inhibits DENV-2 infection and provides guidance for antiviral treatment strategies for DENV infections.


Asunto(s)
Virus del Dengue , Dengue , Animales , Barrera Hematoencefálica , Virus del Dengue/fisiología , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Vimentina/metabolismo
2.
Microb Drug Resist ; 28(1): 7-17, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34357802

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen, which usually presents multiple antibiotic resistance. Host-directed therapy involves modulating the host defense system and the interplay between innate and adaptive immunity is a new strategy for designing anti-infection drugs. Memantine (MEM), a drug used to treat Alzheimer's disease, has a good inhibitory effect on neonatal mice with Escherichia coli-associated bacteremia and meningitis; however, the inhibitory effect and mechanisms of MEM against P. aeruginosa infection remain unclear. Here, we investigated whether MEM could inhibit P. aeruginosa infection and explored the potential mechanisms. MEM significantly promoted the bactericidal effect of neutrophils against P. aeruginosa and its drug-resistant strain. The combination index of MEM and amikacin (AMK) was <1. In vivo experiments showed that the bacteremia and inflammation severities in the MEM-treated group were less than those in the untreated group, and the bacterial load in the organs was significantly less than that in the control group. Combining MEM with the reactive oxygen species (ROS) inhibitor, N-acetyl-l-cysteine, weakened the anti-infective effect of MEM. MEM increased the expression of NADPH p67phox and promoted neutrophilic ROS production. Deleting the p67phox gene significantly weakened the effects of MEM on ROS generation and improving bactericidal effect of neutrophils. In conclusion, MEM promoted the bactericidal effect of neutrophils against P. aeruginosa and its drug-resistant strain, and had a synergistic antibacterial effect when combined with AMK. MEM may exert its anti-infective effects by promoting neutrophilic bactericidal activity via increasing the expression level of p67phox and further stimulating ROS generation.


Asunto(s)
Amicacina/farmacología , Antibacterianos/farmacología , Memantina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Farmacorresistencia Bacteriana , Neutrófilos/efectos de los fármacos , Fosfoproteínas/efectos de los fármacos , Infecciones por Pseudomonas/prevención & control , Ratas , Ratas Sprague-Dawley
3.
Front Immunol ; 12: 745854, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721415

RESUMEN

Alpha 7 nicotinic acetylcholine receptor (α7 nAChR) is critical for the pathogenesis of Escherichia coli (E. coli) K1 meningitis, a severe central nervous system infection of the neonates. However, little is known about how E. coli K1 manipulates α7 nAChR signaling. Here, through employing immortalized cell lines, animal models, and human transcriptional analysis, we showed that E. coli K1 infection triggers releasing of secreted Ly6/Plaur domain containing 1 (SLURP1), an endogenous α7 nAChR ligand. Exogenous supplement of SLURP1, combined with SLURP1 knockdown or overexpression cell lines, showed that SLURP1 is required for E. coli K1 invasion and neutrophils migrating across the blood-brain barrier (BBB). Furthermore, we found that SLURP1 is required for E. coli K1-induced α7 nAChR activation. Finally, the promoting effects of SLURP1 on the pathogenesis of E. coli K1 meningitis was significantly abolished in the α7 nAChR knockout mice. These results reveal that E. coli K1 exploits SLURP1 to activate α7 nAChR and facilitate its pathogenesis, and blocking SLURP1-α7 nAChR interaction might represent a novel therapeutic strategy for E. coli K1 meningitis.


Asunto(s)
Antígenos Ly/fisiología , Barrera Hematoencefálica , Infecciones por Escherichia coli/microbiología , Escherichia coli/fisiología , Meningitis por Escherichia coli/fisiopatología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Antígenos Ly/genética , Línea Celular , Líquido Cefalorraquídeo/microbiología , Células Endoteliales/microbiología , Escherichia coli/aislamiento & purificación , Hipocampo/metabolismo , Interacciones Huésped-Patógeno , Humanos , Recién Nacido , Memantina/farmacología , Meningitis por Escherichia coli/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/fisiología , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Activador de Plasminógeno de Tipo Uroquinasa/genética , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores , Receptor Nicotínico de Acetilcolina alfa 7/deficiencia
4.
Artículo en Inglés | MEDLINE | ID: mdl-32117815

RESUMEN

Bacterial infection remains one of the leading causes of death worldwide due to the continuous rise of multiple antibiotic-resistant bacteria. Focusing solely on bacteria as the drug targets is a major limitation inherent in the conventional antibiotic therapy. Recently, host-directed therapies have become such an innovative approach to modulate the host defense system and the interplay of innate and adaptive immunity. Our previous studies showed that memantine (MEM), an α7 nAChR antagonist, could efficiently block multi-drug resistant Escherichia coli-caused bacteremia and meningitis in a mouse model. However, the underlying mechanisms that govern the antibacterial effects of MEM are still unknown. In this study, we demonstrated that MEM is able to significantly suppress E. coli infection by enhancing E. coli-induced formation and release of NETs in vitro and in vivo. MEM could promote the trapping and bactericidal activities of the polymorphonuclear neutrophils (PMNs) in a manner dependent on α7 nAChR, since knockdown of this receptor noticeably reduces the survival ability of bacteria in PMNs while MEM no longer affects the survival of bacteria in PMNs. Our results also showed that when the expression of S100A9, an antiseptic protein, is inhibited, pathogen survival rates in PMNs increase significantly. MEM reverses this effect in a concentration-dependent manner. MEM stimulates the production of MPO, S100A9, and DNA in PMNs and accelerates the release of depolymerized chromatin fibers into the extracellular space, suggesting the formation of NETs. Taken together, our data suggest that MEM effectively blocks bacterial infection through the promotion of the antibacterial function of NETs induced by E. coli.


Asunto(s)
Trampas Extracelulares , Meningitis , Animales , Escherichia coli , Memantina/farmacología , Ratones , Neutrófilos
5.
Front Microbiol ; 10: 477, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30923519

RESUMEN

It has long been known that probiotics can be used to maintain intestinal homeostasis and treat a number of gastrointestinal disorders, but the underlying mechanism has remained obscure. Recently, increasing evidence supports the notion that certain probiotic-derived components, such as bacteriocins, lipoteichoic acids, surface layer protein and secreted protein, have a similar protective role on intestinal barrier function as that of live probiotics. These bioactive components have been named 'postbiotics' in the most recent publications. We previously found that the Lactobacillus rhamnosus GG (LGG) culture supernatant is able to accelerate the maturation of neonatal intestinal defense and prevent neonatal rats from oral Escherichia coli K1 infection. However, the identity of the bioactive constituents has not yet been determined. In this study, using liquid chromatography-tandem mass spectrometry analysis, we identified a novel secreted protein (named HM0539 here) involved in the beneficial effect of LGG culture supernatant. HM0539 was recombinated, purified, and applied for exploring its potential bioactivity in vitro and in vivo. Our results showed that HM0539 exhibits a potent protective effect on the intestinal barrier, as reflected by enhancing intestinal mucin expression and preventing against lipopolysaccharide (LPS)- or tumor necrosis factor α (TNF-α)-induced intestinal barrier injury, including downregulation of intestinal mucin (MUC2), zonula occludens-1 (ZO-1) and disruption of the intestinal integrity. Using a neonatal rat model of E. coli K1 infection via the oral route, we verified that HM0539 is sufficient to promote development of neonatal intestinal defense and prevent against E. coli K1 pathogenesis. Moreover, we further extended the role of HM0539 and found it has potential to prevent dextran sulfate sodium (DSS)-induced colitis as well as LPS/D-galactosamine-induced bacterial translocation and liver injury. In conclusion, we identified a novel LGG postbiotic HM0539 which exerts a protective effect on intestinal barrier function. Our findings indicated that HM0539 has potential to become a useful agent for prevention and treatment of intestinal barrier dysfunction- related diseases.

6.
Front Microbiol ; 8: 1798, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28979247

RESUMEN

Escherichia coli (E. coli) K1 sepsis and meningitis is a severe infection characterized by high mortality in neonates. Successful colonization and translocation across the intestinal mucosa have been regarded as the critical steps for E. coli K1 sepsis and meningitis. We recently reported that the probiotic mixture, Golden Bifido (containing live Lactobacillus bulgaricus, Bifidobacterium, and Streptococcus thermophilus, LBS) has a preventive role against neonatal E. coli K1 bacteremia and meningitis. However, the interaction between the neonatal gut barrier, probiotics and E. coli K1 is still not elucidated. The present study aims to investigate how LBS exerts its protective effects on neonatal gut barrier during E. coli K1 infection. The beneficial effects of LBS were explored in vitro and in vivo using human colon carcinoma cell lines HT-29 and rat model of neonatal E. coli K1 infection, respectively. Our results showed that stimulation with E. coli K1 was able to cause intestinal barrier dysfunction, which were reflected by E. coli K1-induced intestinal damage and apoptosis of intestinal epithelial cells, reduction of mucin, immunoglobulin A (IgA) and tight junction proteins expression, as well as increase in intestinal permeability, all these changes facilitate E. coli K1 intestinal translocation. However, these changes were alleviated when HT-29 cells were treated with LBS before E. coli K1 infection. Furthermore, we found that LBS-treated neonatal rats (without E. coli K1 infection) have showed higher production of mucin, ZO-1, IgA, Ki67 in intestinal mucosa as well as lower intestinal permeability than that of non-treated rats, indicating that LBS could accelerate the development of neonatal intestinal defense. Taken together, our results suggest that enhancement of the neonatal intestinal defense to fight against E. coli K1 translocation could be the potential mechanism to elucidate how LBS confers a protective effect against neonatal E. coli K1 bacteremia and meningitis. This indirect mechanism makes LBS exert preventive effect on most of gut-derived pathogenic infections rather than only E. coli.

7.
Sci Rep ; 7: 43305, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28262688

RESUMEN

The objective of this study was to determine whether Lactobacillus rhamnosus GG culture supernatant (LCS) has a preventive effect against gut-derived systemic neonatal Escherichia coli (E. coli) K1 infection. The preventive effects were evaluated in human colonic carcinoma cell line Caco-2 and neonatal rat models. Our in vitro results showed that LCS could block adhesion, invasion and translocation of E. coli K1 to Caco-2 monolayer via up-regulating mucin production and maintaining intestinal integrity. In vivo experiments revealed that pre-treatment with LCS significantly decrease susceptibility of neonatal rats to oral E. coli K1 infection as reflected by reduced bacterial intestinal colonization, translocation, dissemination and systemic infections. Further, we found that LCS treated neonatal rats have higher intestinal expressions of Ki67, MUC2, ZO-1, IgA, mucin and lower barrier permeability than those in untreated rats. These results indicated that LCS could enhance neonatal resistance to systemic E. coli K1 infection via promoting maturation of neonatal intestinal defense. In conclusions, our findings suggested that LCS has a prophylactic effect against systemic E. coli K1 infection in neonates. Future studies aimed at identifying the specific active ingredients in LCS will be helpful in developing effective pharmacological strategies for preventing neonatal E. coli K1 infection.


Asunto(s)
Antibacterianos/metabolismo , Infecciones por Escherichia coli/prevención & control , Escherichia coli/fisiología , Lacticaseibacillus rhamnosus/metabolismo , Sepsis Neonatal/prevención & control , Animales , Animales Recién Nacidos , Antígenos Bacterianos/análisis , Adhesión Bacteriana/efectos de los fármacos , Traslocación Bacteriana/efectos de los fármacos , Células CACO-2 , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Humanos , Polisacáridos Bacterianos/análisis , Ratas
8.
Sci Rep ; 7: 40467, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28074940

RESUMEN

One of the most challenging issues in HIV-associated neurocognitive disorders (HAND) caused by HIV-1 virotoxins and drug abuse is the lack of understanding the underlying mechanisms that are commonly associated with disorders of the blood-brain barrier (BBB), which mainly consists of brain microvascular endothelial cells (BMEC). Here, we hypothesized that Glycoprotein 120 (gp120), methamphetamine (METH) and nicotine (NT) can enhance amyloid-beta (Aß) accumulation in BMEC through Alpha7 nicotinic acetylcholine receptor (α7 nAChR). Both in vitro (human BMEC) (HBMEC) and in vivo (mice) models of BBB were used to dissect the role of α7 nAChR in up-regulation of Aß induced by gp120, METH and NT. Aß release from and transport across HBMEC were significantly increased by these factors. Methyllycaconitine (MLA), an antagonist of α7 nAChR, could efficiently block these pathogenic effects. Furthermore, our animal data showed that these factors could significantly increase the levels of Aß, Tau and Ubiquitin C-Terminal Hydrolase L1 (UCHL1) in mouse cerebrospinal fluid (CSF) and Aß in the mouse brains. These pathogenicities were significantly reduced by MLA, suggesting that α7 nAChR may play an important role in neuropathology caused by gp120, METH and NT, which are the major pathogenic factors contributing to the pathogenesis of HAND.


Asunto(s)
Amiloide/metabolismo , Encéfalo/patología , Células Endoteliales/metabolismo , Proteína gp120 de Envoltorio del VIH/farmacología , Metanfetamina/farmacología , Nicotina/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Aconitina/análogos & derivados , Aconitina/farmacología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Biomarcadores/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/lesiones , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Movimiento Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células HL-60 , Humanos , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas S100/metabolismo , Factores de Tiempo , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(1): 24-29, 2017 01 20.
Artículo en Chino | MEDLINE | ID: mdl-28109094

RESUMEN

OBJECTIVE: To investigate whether Lactobacillus rhamnosus GG conditioned medium(LGG-CM)has preventive effect against E. coli K1-induced neuropathogenicity in vitro by inhibiting nuclear factor-κB (NF-κB) signaling pathway. METHODS: An in vitro blood-brain barrier (BBB) model was constructed using human brain microvascular endothelial cells (HBMECs). The effect of LGG-CM on E. coli-actived NF-κB signaling pathway was assayed using Western blotting. Invasion assay and polymorphonuclear leukocyte (PMN) transmigration assay were performed to explore whether LGG-CM could inhibit E. coli invasion and PMN transmigration across the BBB in vitro. The expressions of ZO-1 and CD44 were detected using Western blotting and immunofluorescence. The changes of trans-epithelial electric resistance (TEER) and bacterial translocation were determined to evaluate the BBB permeability. RESULTS: Pre-treament with LGG-CM inhibited E. coli-activated NF-κB signaling pathway in HBMECs and decreased the invasion of E. coli K1 and transmigration of PMN. Western blotting showed that LGG-CM could alleviate E. coli-induced up-regulation of CD44 and down-regulation of ZO-1 expressions in HBMECs. In addition, pre-treatment with LGG-CM alleviated E. coli K1-induced reduction of TEER and suppressed bacterial translocation across the BBB in vitro. CONCLUSION: LGG-CM can block E. coli-induced activation of NF-κB signaling pathway and thereby prevents E. coli K1-induced neuropathogenicity by decreasing E. coli K1 invasion rates and PMN transmigration.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Escherichia coli/efectos de los fármacos , Lacticaseibacillus rhamnosus , Meningitis por Escherichia coli/prevención & control , FN-kappa B/antagonistas & inhibidores , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Barrera Hematoencefálica , Escherichia coli/fisiología , Humanos , FN-kappa B/metabolismo , Neutrófilos/fisiología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/fisiología
10.
Curr Top Med Chem ; 17(3): 278-289, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27572078

RESUMEN

High lethality of infections caused by Listeria monocytogenes still remains a major clinical problem in spite of their susceptibility to a wide spectrum of antibiotics. The refractoriness towards treatment is primarily due to its amazing capacity to invade non-phagocytic cells and replicate there in, imparting the dual protection from immune response and antimicrobials. Therefore, generating new anti-infective drugs against intracellular infections has emerged as an urgent issue in the therapeutics of listeriosis. Researches have demonstrated that, internalization of Listeria monocytogenes into nonphagocytic cells is mediated by the interactions between the two bacterial invasion proteins, InlA and InlB, and their cellular surface receptors, E-cadherin and c-Met. As InlB promotes entry into various cell types, such as hepatocytes, epithelial cells and endothelial cells, targeting of InlB-c-Met mediated invasion is important for specifically blocking their intracellular infection. Furthermore, our preliminary in vitro studies have shown that a GA (Geldanamycin, GA) analogue, 17-AAG (tanespimycin) which is widely used in cancer therapy have important therapeutic potential by significantly enhancing the capacity of ampicillin to kill intracellular L. monocytogenes, and to protect the infected HBMECs from the cytocidal effects of this bacterium. We report here, the feasibility of tanespimycin as a potential anti-intracellular infective drug and its clinical relevance in a broader prospective, including the significant advancements in therapeutic approaches, drug effectiveness and toxicity. Exploring the therapeutic effects of c-Met inhibitors such as tanespimycin on L. monocytogenes intracellular infection may provide an alternative novel strategy for the development of antimicrobial agents for treatment of infectious diseases.


Asunto(s)
Antibacterianos/uso terapéutico , Listeria monocytogenes/efectos de los fármacos , Listeriosis/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Humanos , Listeria monocytogenes/patogenicidad , Listeria monocytogenes/fisiología , Virulencia
12.
PLoS One ; 11(9): e0162641, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27657497

RESUMEN

BACKGROUND: NF-κB activation, pathogen invasion, polymorphonuclear leukocytes (PMN) transmigration (PMNT) across the blood-brain barrier (BBB) are the pathogenic triad hallmark features of bacterial meningitis, but the mechanisms underlying these events remain largely unknown. Vimentin, which is a novel NF-κB regulator, is the primary receptor for the major Escherichia coli K1 virulence factor IbeA that contributes to the pathogenesis of neonatal bacterial sepsis and meningitis (NSM). We have previously shown that IbeA-induced NF-κB signaling through its primary receptor vimentin as well as its co-receptor PTB-associated splicing factor (PSF) is required for pathogen penetration and leukocyte transmigration across the BBB. This is the first in vivo study to demonstrate how vimentin and related factors contributed to the pathogenic triad of bacterial meningitis. METHODOLOGY/PRINCIPAL FINDINGS: The role of vimentin in IbeA+ E. coli K1-induced NF-κB activation, pathogen invasion, leukocyte transmigration across the BBB has now been demonstrated by using vimentin knockout (KO) mice. In the in vivo studies presented here, IbeA-induced NF-κB activation, E. coli K1 invasion and polymorphonuclear neutrophil (PMN) transmigration across the BBB were significantly reduced in Vim-/- mice. Decreased neuronal injury in the hippocampal dentate gyrus was observed in Vim-/- mice with meningitis. The major inflammatory regulator α7 nAChR and several signaling molecules contributing to NF-κB activation (p65 and p-CamKII) were significantly reduced in the brain tissues of the Vim-/- mice with E. coli meningitis. Furthermore, Vim KO resulted in significant reduction in neuronal injury and in α7 nAChR-mediated calcium signaling. CONCLUSION/SIGNIFICANCE: Vimentin, a novel NF-κB regulator, plays a detrimental role in the host defense against meningitic infection by modulating the NF-κB signaling pathway to increase pathogen invasion, PMN recruitment, BBB permeability and neuronal inflammation. Our findings provide the first evidence for Vim-dependent mechanisms underlying the pathogenic triad of bacterial meningitis.

13.
PLoS One ; 11(6): e0156170, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27280726

RESUMEN

To explore the differences between the extreme SIV infection phenotypes, nonprogression (BEN: benign) to AIDS in sooty mangabeys (SMs) and progression to AIDS (MAL: malignant) in rhesus macaques (RMs), we performed an integrated dual positive-negative connectivity (DPNC) analysis of gene coexpression networks (GCN) based on publicly available big data sets in the GEO database of NCBI. The microarray-based gene expression data sets were generated, respectively, from the peripheral blood of SMs and RMs at several time points of SIV infection. Significant differences of GCN changes in DPNC values were observed in SIV-infected SMs and RMs. There are three groups of enriched genes or pathways (EGPs) that are associated with three SIV infection phenotypes (BEN+, MAL+ and mixed BEN+/MAL+). The MAL+ phenotype in SIV-infected RMs is specifically associated with eight EGPs, including the protein ubiquitin proteasome system, p53, granzyme A, gramzyme B, polo-like kinase, Glucocorticoid receptor, oxidative phosyphorylation and mitochondrial signaling. Mitochondrial (endosymbiotic) dysfunction is solely present in RMs. Specific BEN+ pattern changes in four EGPs are identified in SIV-infected SMs, including the pathways contributing to interferon signaling, BRCA1/DNA damage response, PKR/INF induction and LGALS8. There are three enriched pathways (PRR-activated IRF signaling, RIG1-like receptor and PRR pathway) contributing to the mixed (BEN+/MAL+) phenotypes of SIV infections in RMs and SMs, suggesting that these pathways play a dual role in the host defense against viral infections. Further analysis of Hub genes in these GCNs revealed that the genes LGALS8 and IL-17RA, which positively regulate the barrier function of the gut mucosa and the immune homeostasis with the gut microbiota (exosymbiosis), were significantly differentially expressed in RMs and SMs. Our data suggest that there exists an exo- (dysbiosis of the gut microbiota) and endo- (mitochondrial dysfunction) symbiotic imbalance (EESI) in HIV/SIV infections. Dissecting the mechanisms of the exo-endo symbiotic balance (EESB) that maintains immune homeostasis and the EESI problems in HIV/SIV infections may lead to a better understanding of the pathogenesis of AIDS and the development of novel interventions for the rational control of this disease.


Asunto(s)
Cercocebus atys/genética , Redes Reguladoras de Genes , Macaca mulatta/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Cercocebus atys/inmunología , Cercocebus atys/virología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Macaca mulatta/inmunología , Macaca mulatta/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Especificidad de la Especie
14.
Front Microbiol ; 7: 148, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925035

RESUMEN

Bacterial meningitis remains the leading cause of disabilities worldwide. This life-threatening disease has a high mortality rate despite the availability of antibiotics and improved critical care. The interactions between bacterial surface components and host defense systems that initiate bacterial meningitis have been studied in molecular and cellular detail over the past several decades. Bacterial meningitis commonly exhibits triad hallmark features (THFs): pathogen penetration, nuclear factor-kappaB (NF-κB) activation in coordination with type 1 interferon (IFN) signaling and leukocyte transmigration that occur at the blood-brain barrier (BBB), which consists mainly of brain microvascular endothelial cells (BMEC). This review outlines the progression of these early inter-correlated events contributing to the central nervous system (CNS) inflammation and injury during the pathogenesis of bacterial meningitis. A better understanding of these issues is not only imperative to elucidating the pathogenic mechanism of bacterial meningitis, but may also provide the in-depth insight into the development of novel therapeutic interventions against this disease.

15.
J Biomed Sci ; 23: 28, 2016 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-26897523

RESUMEN

BACKGROUND: Cryptococcus neoformans (Cn) is an important opportunistic pathogen in the immunocompromised people, including AIDS patients, which leads to fatal cryptococcal meningitis with high mortality rate. Previous researches have shown that HIV-1 gp41-I90 ectodomain can enhance Cn adhesion to and invasion of brain microvascular endothelial cell (BMEC), which constitutes the blood brain barrier (BBB). However, little is known about the role of HIV-1 gp41-I90 in the monocyte transmigration across Cn-infected BBB. In the present study, we provide evidence that HIV-1 gp41-I90 and Cn synergistically enhance monocytes transmigration across the BBB in vitro and in vivo. The underlying mechanisms for this phenomenon require further study. METHODS: In this study, the enhancing role of HIV-1 gp41-I90 in monocyte transmigration across Cn-infected BBB was demonstrated by performed transmigration assays in vitro and in vivo. RESULTS: Our results showed that the transmigration rate of monocytes are positively associated with Cn and/or HIV-1 gp41-I90, the co-exposure (HIV-1 gp41-I90 + Cn) group showed a higher THP-1 transmigration rate (P < 0.01). Using CD44 knock-down HBMEC or CD44 inhibitor Bikunin in the assay, the facilitation of transmigration rates of monocyte enhanced by HIV-1 gp41-I90 was significantly suppressed. Western blotting analysis and biotin/avidin enzyme-linked immunosorbent assays (BA-ELISAs) showed that Cn and HIV-1 gp41-I90 could increase the expression of CD44 and ICAM-1 on the HBMEC. Moreover, Cn and/or HIV-1 gp41-I90 could also induce CD44 redistribution to the membrane lipid rafts. By establishing the mouse cryptococcal meningitis model, we found that HIV-1 gp41-I90 and Cn could synergistically enhance the monocytes transmigration, increase the BBB permeability and injury in vivo. CONCLUSIONS: Collectively, our findings suggested that HIV-1 gp41-I90 ectodomain can enhance the transmigration of THP-1 through Cn-infected BBB, which may be mediated by CD44. This novel study enlightens the future prospects to elaborate the inflammatory responses induced by HIV-1 gp41-I90 ectodomain and to effectively eliminate the opportunistic infections in AIDS patients.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Criptococosis/metabolismo , Cryptococcus neoformans , Células Endoteliales/metabolismo , Proteína gp41 de Envoltorio del VIH/metabolismo , VIH-1 , Receptores de Hialuranos/metabolismo , Monocitos/metabolismo , Migración Transendotelial y Transepitelial , Animales , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/virología , Línea Celular , Criptococosis/genética , Células Endoteliales/microbiología , Células Endoteliales/virología , Proteína gp41 de Envoltorio del VIH/genética , Humanos , Receptores de Hialuranos/genética , Ratones , Ratones Noqueados , Estructura Terciaria de Proteína
16.
J Microbiol Biotechnol ; 26(5): 918-27, 2016 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26437944

RESUMEN

Cryptococcus neoformans is a life-threatening pathogenic yeast that causes devastating meningoencephalitis. The mechanism of cryptococcal brain invasion is largely unknown, and recent studies suggest that its extracellular microvesicles may be involved in the invasion process. The 14-3-3 protein is abundant in the extracellular microvesicles of C. neoformans, and the 14-3-3-GFP fusion has been used as the microvesicle's marker. However, the physiological role of 14-3-3 has not been explored. In this report, we have found that C. neoformans contains a single 14-3-3 gene that apparently is an essential gene. To explore the functions of 14-3-3, we substituted the promoter region of the 14-3-3 with the copper-controllable promoter CTR4. The CTR4 regulatory strain showed an enlarged cell size, drastic changes in morphology, and a decrease in the thickness of the capsule under copper-enriched conditions. Furthermore, the mutant cells produced a lower amount of total proteins in their extracellular microvesicles and reduced adhesion to human brain microvascular endothelial cells in vitro. Proteomic analyses of the protein components under 14-3-3-overexpressed and -suppressed conditions revealed that the 14-3-3 function(s) might be associated with the microvesicle biogenesis. Our results support that 14-3-3 has diverse pertinent roles in both physiology and pathogenesis in C. neoformans. Its gene functions are closely relevant to the pathogenesis of this fungus.


Asunto(s)
Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Cryptococcus neoformans/genética , Fosfatasa Ácida/metabolismo , Biomarcadores/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/microbiología , Adhesión Celular/fisiología , Cobre/metabolismo , Cryptococcus neoformans/crecimiento & desarrollo , Cryptococcus neoformans/patogenicidad , Células Endoteliales/microbiología , Cápsulas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Lacasa/metabolismo , Mutación , Fenotipo , Regiones Promotoras Genéticas , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
17.
Med Microbiol Immunol ; 205(2): 97-109, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26233310

RESUMEN

Proteus mirabilis (P. mirabilis), a gram-negative enteric bacterium, frequently causes urinary tract infections. Many virulence factors of uropathogenic P. mirabilis have been identified, including urease, flagella, hemolysin and fimbriae. However, the functions of polyphosphate kinase (PPK), which are related to the pathogenicity of many bacteria, remain entirely unknown in P. mirabilis. In this study, a ppk gene encoding the PPK insertional mutant in P. mirabilis strain HI4320 was constructed, and its biological functions were examined. The results of survival studies demonstrated that the ppk mutant was deficient in resistance to oxidative, hyperosmotic and heat stress. The swarming and biofilm formation abilities of P. mirabilis were also attenuated after the ppk interruption. In vitro and in vivo experiments suggested that ppk was required for P. mirabilis to invade the bladder. The negative phenotypes of the ppk mutant could be restored by ppk gene complementation. Furthermore, two-dimensional gel electrophoresis and liquid chromatography-mass spectrometry were used to analyze the proteomes of the wild-type strain and the ppk mutant. Compared with the wild-type strain, seven proteins including TonB-dependent receptor, universal stress protein G, major mannose-resistant/Proteus-like fimbrial protein (MR/P fimbriae), heat shock protein, flagellar capping protein, putative membrane protein and multidrug efflux protein were down-regulated, and four proteins including exported peptidase, repressor protein for FtsI, FKBP-type peptidyl-prolyl cis-trans isomerase and phosphotransferase were up-regulated in the ppk mutant. As a whole, these results indicate that PPK is an important regulator and plays a crucial role in stress tolerance and virulence in uropathogenic P. mirabilis.


Asunto(s)
Adaptación Biológica , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Infecciones por Proteus/microbiología , Proteus mirabilis/fisiología , Estrés Fisiológico , Adaptación Biológica/genética , Animales , Adhesión Bacteriana/genética , Carga Bacteriana , Biopelículas , Modelos Animales de Enfermedad , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , Mutación , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Infecciones Urinarias/microbiología , Virulencia/genética , Factores de Virulencia/genética
19.
BMC Infect Dis ; 15: 352, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26285576

RESUMEN

BACKGROUND: Cryptococcal meningitis is the most common fungal infection of the central nervous system (CNS) in HIV/AIDS. HIV-1 virotoxins (e.g., gp41) are able to induce disorders of the blood-brain barrier (BBB), which mainly consists of BMEC. Our recent study suggests that α7 nAChR is an essential regulator of inflammation, which contributes to regulation of NF-κB signaling, neuroinflammation and BBB disorders caused by microbial (e.g., HIV-1 gp120) and non-microbial [e.g., methamphetamine (METH)] factors. However, the underlying mechanisms for multiple comorbidities are unclear. METHODS: In this report, an aggravating role of α7 nAChR in host defense against CNS disorders caused by these comorbidities was demonstrated by chemical [inhibitor: methyllycaconitine (MLA)] and genetic (α7(-/-) mice) blockages of α7 nAChR. RESULTS: As shown in our in vivo studies, BBB injury was significantly reduced in α7(-/-) mice infected with C. neoformans. Stimulation by the gp41 ectodomain peptide (gp41-I90) and METH was abolished in the α7(-/-) animals. C. neoformans and gp41-I90 could activate NF-κB. Gp41-I90- and METH-induced monocyte transmigration and senescence were significantly inhibited by MLA and CAPE (caffeic acid phenethyl ester, an NF-κB inhibitor). CONCLUSIONS: Collectively, our data suggest that α7 nAChR plays a detrimental role in the host defense against C. neoformans- and HIV-1 associated comorbidity factors-induced BBB injury and CNS disorders.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Cryptococcus neoformans , Meningitis Criptocócica/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Aconitina/análogos & derivados , Aconitina/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Coinfección , Proteína gp41 de Envoltorio del VIH/farmacología , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Inflamación , Metanfetamina/farmacología , Ratones , Ratones Noqueados , FN-kappa B/efectos de los fármacos , Antagonistas Nicotínicos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores
20.
J Microbiol Biotechnol ; 25(10): 1751-60, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26059517

RESUMEN

Mucin2 (MUC2), an important regulatory factor in the immune system, plays an important role in the host defense system against bacterial translocation. Probiotics known to regulate MUC2 gene expression have been widely studied, but the interactions among probiotic, pathogens, and mucin gene are still not fully understood. The aim of this study was to investigate the role of MUC2 in blocking effects of probiotics on meningitic E. coli-induced pathogenicities. In this study, live combined probiotic tablets containing living Bifidobacterium, Lactobacillus bulgaricus, and Streptococcus thermophilus were used. MUC2 expression was knocked down in Caco-2 cells by RNA interference. 5-Aza-2'-deoxycytidine (5-Aza-CdR), which enhances mucin-promoted probiotic effects through inducing production of Sadenosyl- L-methionine (SAMe), was used to up-regulate MUC2 expression in Caco-2 cells. The adhesion to and invasion of meningitic E. coli were detected by competition assays. Our studies showed that probiotic agents could block E. coli-caused intestinal colonization, bacteremia, and meningitis in a neonatal sepsis and meningitis rat model. MUC2 gene expression in the neonatal rats given probiotic agents was obviously higher than that of the infected and uninfected control groups without probiotic treatment. The prohibitive effects of probiotic agents on MUC2-knockdown Caco-2 cells infected with E44 were significantly reduced compared with nontransfected Caco-2 cells. Moreover, the results also showed that 5- Aza-CdR, a drug enhancing the production of SAMe that is a protective agent of probiotics, was able to significantly suppress adhesion and invasion of E44 to Caco-2 cells by upregulation of MUC2 expression. Taken together, our data suggest that probiotic agents can efficiently block meningitic E. coli-induced pathogenicities in a manner dependent on MUC2.


Asunto(s)
Antibiosis , Bifidobacterium/inmunología , Escherichia coli/inmunología , Lactobacillus/inmunología , Mucina 2/metabolismo , Probióticos/farmacología , Streptococcus thermophilus/inmunología , Animales , Animales Recién Nacidos , Bifidobacterium/fisiología , Células CACO-2 , Modelos Animales de Enfermedad , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/prevención & control , Humanos , Lactobacillus/fisiología , Meningitis Bacterianas/prevención & control , Modelos Biológicos , Ratas , Sepsis/prevención & control , Streptococcus thermophilus/fisiología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA