Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancer Cell Int ; 23(1): 107, 2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37268941

RESUMEN

BACKGROUND: Radiotherapy is one of the effective methods for treatment of breast cancer; however, controversies still exist with respect to radiotherapy for patients with TNBC. Here, we intend to explore the mechanism by which local radiotherapy promotes the recruitment of M-MDSCs in the lung and increases the risk of lung metastasis in TNBC tumor-bearing mice. METHODS: A single dose of 20 Gy X-ray was used to locally irradiate the primary tumor of 4T1 tumor-bearing mice. Tumor growth, the number of pulmonary metastatic nodules, and the frequency of MDSCs were monitored in the mice. Antibody microarray and ELISA methods were used to analyze the cytokines in exosomes released by irradiated (IR) or non-IR 4T1 cells. The effects of the exosomes on recruitment of MDSCs and colonization of 4T1 cells in the lung of normal BALB/c mice were observed with the methods of FCM and pathological section staining. T lymphocytes or 4T1 cells co-cultured with MDSCs were performed to demonstrate the inhibitory effect on T lymphocytes or accelerative migration effect on 4T1 cells. Finally, a series of in vitro experiments demonstrated how the exosomes promote the recruitment of M-MDSCs in lung of mice. RESULTS: Even though radiotherapy reduced the burden of primary tumors and larger lung metastatic nodules (≥ 0.4 mm2), the number of smaller metastases (< 0.4 mm2) significantly increased. Consistently, radiotherapy markedly potentiated M-MDSCs and decreased PMN-MDSCs recruitment to lung of tumor-bearing mice. Moreover, the frequency of M-MDSCs of lung was positively correlated with the number of lung metastatic nodules. Further, M-MDSCs markedly inhibited T cell function, while there was no difference between M-MDSCs and PMN-MDSCs in promoting 4T1 cell migration. X-ray irradiation promoted the release of G-CSF, GM-CSF and CXCl1-rich exosomes, and facilitated the migration of M-MDSCs and PMN-MDSCs into the lung through CXCL1/CXCR2 signaling. While irradiated mouse lung extracts or ir/4T1-exo treated macrophage culture medium showed obvious selective chemotaxis to M-MDSCs. Mechanistically, ir/4T1-exo induce macrophage to produce GM-CSF, which further promoted CCL2 release in an autocrine manner to recruit M-MDSCs via CCL2/CCR2 axis. CONCLUSIONS: Our work has identified an undesired effect of radiotherapy that may promote immunosuppressive premetastatic niches formation by recruiting M-MDSCs to lung. Further studies on radiotherapy combined CXCR2 or CCR2 signals inhibitors were necessary.

2.
Front Immunol ; 12: 654998, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531848

RESUMEN

HCV core protein is the first structural protein synthesized during hepatitis C virus (HCV) infection and replication. It is released from virus infected liver cells and mediates multiple functions to affect host cell response. The innate immune response is the first line of defense against viral infection. After HCV infection, Kupffer cells (KCs) which are liver macrophages play an important role in host innate immune response. Kupffer cells act as phagocytes and release different cytokines and chemokines to counter viral infection and regulate inflammation and fibrosis in liver. Earlier, we have demonstrated that HCV core protein interacts with gC1qR and activates MAPK, NF-κB and PI3K/AKT pathways in macrophages. In this study, we explored the effect of HCV core protein on CCL2 and CXCL10 expression in macrophages and the signaling pathways involved. Upon silencing of gC1qR, we observed a significant decrease expression of CCL2 and CXCL10 in macrophages in the presence of HCV core protein. Inhibiting NF-κB pathway, but not P38, JNK, ERK and AKT pathways greatly reduced the expression of CCL2 and CXCL10. Therefore, our results indicate that interaction of HCV core protein with gC1qR could induce CCL2 and CXCL10 secretion in macrophages via NF-κB signaling pathway. These findings may shed light on the understanding of how leukocytes migrate into the liver and exaggerate host-derived immune responses and may provide novel therapeutic targets in HCV chronic inflammation.


Asunto(s)
Quimiocina CCL2/inmunología , Quimiocina CXCL10/inmunología , Hepacivirus/inmunología , Macrófagos/inmunología , FN-kappa B/inmunología , Transducción de Señal/inmunología , Proteínas del Núcleo Viral/inmunología , Animales , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Expresión Génica/inmunología , Hepacivirus/metabolismo , Hepacivirus/fisiología , Hepatitis C/inmunología , Hepatitis C/metabolismo , Hepatitis C/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/virología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Células THP-1 , Proteínas del Núcleo Viral/metabolismo
3.
Front Cell Infect Microbiol ; 11: 628275, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33791238

RESUMEN

The Tripartite motif (TRIM) protein family, which contains over 80 members in human sapiens, is the largest subfamily of the RING-type E3 ubiquitin ligase family. It is implicated in regulating various cellular functions, including cell cycle process, autophagy, and immune response. The dysfunction of TRIMs may lead to numerous diseases, such as systemic lupus erythematosus (SLE). Lots of studies in recent years have demonstrated that many TRIM proteins exert antiviral roles. TRIM proteins could affect viral replication by regulating the signaling pathways of antiviral innate immune responses. Besides, TRIM proteins can directly target viral components, which can lead to the degradation or functional inhibition of viral protein through degradative or non-degradative mechanisms and consequently interrupt the viral lifecycle. However, new evidence suggests that some viruses may manipulate TRIM proteins for their replication. Here, we summarize the latest discoveries on the interactions between TRIM protein and virus, especially TRIM proteins' role in the signaling pathway of antiviral innate immune response and the direct "game" between them.


Asunto(s)
Antivirales , Virus , Humanos , Inmunidad Innata , Transducción de Señal , Proteínas de Motivos Tripartitos
4.
Front Immunol ; 12: 800630, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003126

RESUMEN

Tumorigenesis is a complex multifactorial and multistep process in which tumors can utilize a diverse repertoire of immunosuppressive mechanisms to evade host immune attacks. The degradation of tryptophan into immunosuppressive kynurenine is considered an important immunosuppressive mechanism in the tumor microenvironment. There are three enzymes, namely, tryptophan 2,3-dioxygenase (TDO), indoleamine 2,3-dioxygenase 1 (IDO1), and indoleamine 2,3-dioxygenase 2 (IDO2), involved in the metabolism of tryptophan. IDO1 has a wider distribution and higher activity in catalyzing tryptophan than the other two; therefore, it has been studied most extensively. IDO1 is a cytosolic monomeric, heme-containing enzyme, which is now considered an authentic immune regulator and represents one of the promising drug targets for tumor immunotherapy. Collectively, this review highlights the regulation of IDO1 gene expression and the ambivalent mechanisms of IDO1 on the antitumoral immune response. Further, new therapeutic targets via the regulation of IDO1 are discussed. A comprehensive analysis of the expression and biological function of IDO1 can help us to understand the therapeutic strategies of the inhibitors targeting IDO1 in malignant tumors.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias/inmunología , Triptófano/metabolismo , Animales , Humanos , Tolerancia Inmunológica , Terapia Molecular Dirigida , Microambiente Tumoral
5.
Artículo en Inglés | MEDLINE | ID: mdl-29868491

RESUMEN

Group A streptococcus (GAS), a common pathogen, is able to escape host immune attack and thus survive for longer periods of time. One of the mechanisms used by GAS is the upregulated expression of immunosuppressive molecules, which leads to a reduction in the production of inflammatory cytokines in immune cells. In the present study, we found that macrophages produced lower levels of proinflammatory cytokines (IL-1ß, TNF-α, IL-6) when challenged with GAS than they did when challenged with Escherichia coli (E. coli). Simultaneously, in a mouse model of lung infection, GAS appeared to induce a weaker inflammatory response compared to E. coli. Our data also indicated that the expression of the A20 transcriptional regulator was higher in GAS-infected macrophages than that in macrophages infected with E. coli, and that high expression of A20 correlated with a reduction in the production of TRAF6. SiRNA targeting of A20 led to the increased production of TRAF6, IL-1ß, TNF-α, and IL-6, suggesting that A20 inhibits synthesis of these key proinflammatory cytokines. We also investigated the pathway underlying A20 production and found that the synthesis of A20 depends on My88, and to a lower extent on TNFR1. Finally, we showed a significant reduction in the expression of A20 in macrophages stimulated by M protein-mutant GAS, however, a speB-GAS mutant, which is unable to degrade M protein, induced a greater level of A20 production than wild type GAS. Collectively, our data suggested that M protein of GAS was responsible for inducing A20 expression in macrophages, which in turn down-regulates the inflammatory cytokine response in order to facilitate GAS in evading immune surveillance and thus prolong survival in the host.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Pulmón/inmunología , Macrófagos/inmunología , Neumonía Neumocócica/metabolismo , Streptococcus pyogenes/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/inmunología , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/microbiología , Células RAW 264.7 , Streptococcus pyogenes/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Virol ; 92(8)2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29386287

RESUMEN

Respiratory syncytial virus (RSV) is the main cause of acute lower respiratory tract infection (ALRI) in children worldwide. Virus-host interactions affect the progression and prognosis of the infection. Autophagy plays important roles in virus-host interactions. Respiratory epithelial cells serve as the front line of host defense during RSV infection, However, it is still unclear how they interact with RSV. In this study, we found that RSV induced autophagy that favored RSV replication and exacerbated lung pathology in vivo Mechanistically, RSV induced complete autophagy flux through reactive oxygen species (ROS) generation and activation of the AMP-activated protein kinase/mammalian target of rapamycin (AMPK-MTOR) signaling pathway in HEp-2 cells. Furthermore, we evaluated the functions of autophagy in RSV replication and found that RSV replication was increased in HEp-2 cells treated with rapamycin but decreased remarkably in cells treated with 3-methylademine (3-MA) or wortmannin. Knockdown key molecules in the autophagy pathway with short hairpinp RNA (shRNA) against autophagy-related gene 5 (ATG5), autophagy-related gene 7 (ATG7), or BECN1/Beclin 1 or treatment with ROS scavenger N-acetyl-l-cysteine (NAC) and AMPK inhibitor (compound C) suppressed RSV replication. 3-MA or shATG5/BECN1 significantly decreased cell viability and increased cell apoptosis at 48 hours postinfection (hpi). Blocking apoptosis with Z-VAD-FMK partially restored virus replication at 48 hpi. Those results provide strong evidence that autophagy may function as a proviral mechanism in a cell-intrinsic manner during RSV infection.IMPORTANCE An understanding of the mechanisms that respiratory syncytial virus utilizes to interact with respiratory epithelial cells is critical to the development of novel antiviral strategies. In this study, we found that RSV induces autophagy through a ROS-AMPK signaling axis, which in turn promotes viral infection. Autophagy favors RSV replication through blocking cell apoptosis at 48 hpi. Mechanistically, RSV induces mitophagy, which maintains mitochondrial homeostasis and therefore decreases cytochrome c release and apoptosis induction. This study provides a novel insight into this virus-host interaction, which may help to exploit new antiviral treatments targeting autophagy processes.


Asunto(s)
Apoptosis , Autofagia , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/fisiología , Replicación Viral , Proteínas Quinasas Activadas por AMP/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/metabolismo , Beclina-1/metabolismo , Línea Celular , Humanos , Infecciones por Virus Sincitial Respiratorio/patología , Serina-Treonina Quinasas TOR/metabolismo
7.
Oncotarget ; 7(23): 33796-808, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27183919

RESUMEN

Hepatitis C virus (HCV) infection is characterized by a strong propensity toward chronicity. During chronic HCV infection, HCV core protein is implicated in deregulating cytokine expression that associates with chronic inflammation. A20 is known as a powerful suppressor in cytokine signaling, in this study, we explored the A20 expression in macrophages induced by HCV core protein and the involved signaling pathways. Results demonstrated that HCV core protein induced A20 expression in macrophages. Silencing A20 significantly enhanced the secretion of IL-6, IL-1ß and TGF-ß1, but not IL-8 and TNF. Additionally, HCV core protein interacted with gC1qR, but not TLR2, TLR3 and TLR4 in pull-down assay. Silencing gC1qR abrogated core-induced A20 expression. Furthermore, HCV core protein activated MAPK, NF-κB and PI3K/AKT pathways in macrophages. Inhibition of P38, JNK and NF-κB but not ERK and AKT activities greatly reduced the A20 expression. In conclusion, the study suggests that HCV core protein ligates gC1qR to induce A20 expression in macrophages via P38, JNK and NF-κB signaling pathways, which leads to a low-grade chronic inflammation during HCV infection. It represents a novel mechanism by which HCV usurps the host for persistence.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Hepacivirus/metabolismo , Hepatitis C Crónica/metabolismo , Macrófagos/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteínas del Núcleo Viral/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/virología , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Macrófagos/virología , Ratones , Proteínas Mitocondriales/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteínas del Núcleo Viral/inmunología
8.
PLoS One ; 9(9): e108278, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25265479

RESUMEN

BACKGROUND: The core protein of hepatitis C virus (HCV) is found in the cytoplasm and nuclei of infected cells, including hepatocytes and other cells in the liver. The core protein could be secreted as well. Resident liver macrophages are dependent on the tissue micro-environment and external stimuli to differentiate M1 and M2 hypotypes with distinct functions, and increased expression of the nuclear transcription factor STAT3 was seen in M2-polarized macrophages. In contrast to proinflammatory M1 macrophages, M2 macrophages serve beneficial roles in chronic inflammation, immunosuppression, and tumorigenesis. METHODS: Monocyte-derived human macrophage line (mTHP-1) was treated with the exogenous HCV core protein. Next, the mTHP-1 culture supernatant or cell pellets were added to culture media of normal human liver cell line (L02). RESULTS: Only the culture supernatant stimulated L02 cells proliferation, which was associated with phosphorylated ERK expression. Core protein activated mTHP-1 cells showed enhanced pro- and anti-inflammatory cytokines secretion, which was accompanied by high expression of phosphorylated NF-κB105 and NF-κB65. However, phosphorylated STAT1, and STAT3, which are normally associated with M1 and M2 macrophage polarization, and cell surface expression of CD206, CD14, CD16, and CD86, were unaltered. A transwell co-culture system showed that only in mTHP-1 co-cultured with L02 in the presence of exogenous core protein, were higher levels of phosphorylated STAT3 and CD206 seen. CONCLUSIONS: We showed L02 cells proliferation was accelerated by the culture supernatant of mTHP-1 cells treated with the exogenous HCV core protein. The exogenous core protein mediated the interaction between macrophages and hepatocytes in co-culture, which enhanced the expression of phosphorylated STAT3 and CD206 in macrophages.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Hepatocitos/fisiología , Macrófagos/fisiología , Proteínas Recombinantes/farmacología , Proteínas del Núcleo Viral/farmacología , Antígeno B7-2/biosíntesis , Línea Celular , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Proteínas Ligadas a GPI/biosíntesis , Hepacivirus/genética , Humanos , Lectinas Tipo C/biosíntesis , Receptores de Lipopolisacáridos/biosíntesis , Receptor de Manosa , Lectinas de Unión a Manosa/biosíntesis , Subunidad p50 de NF-kappa B/biosíntesis , Fosforilación , Receptores de Superficie Celular/biosíntesis , Receptores de IgG/biosíntesis , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción ReIA/biosíntesis
9.
PLoS One ; 5(12): e14223, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21151974

RESUMEN

BACKGROUND: Innate immunity of which Toll-like receptor (TLR) 4 and CXCR1 are key elements plays a central role in the development of urinary tract infection (UTI). Although the relation between the genetics of TLR4 and CXCR1 and UTI is investigated partly, the polymorphisms and expression of TLR4 and CXCR1 in different types of UTI in adults are not extremely clear. METHODOLOGY/PRINCIPAL FINDINGS: This study investigates the presence of TLR4 A (896) G and CXCR1 G (2608) C polymorphisms in 129 UTI patients using RFLP-PCR. Gene and allelic prevalence were compared with 248 healthy controls. Flow cytometry was used to detect TLR4 and CXCR1 expression in the monocytes of UTI patients and healthy controls. TLR4 (896) AG genotype and TLR4 (896) G allele had higher prevalence in UTI (especially in acute cystitis and urethritis) patients, whereas CXCR1 (2608) GC genotype and CXCR1 (2608) C allele had lower prevalence in UTI patients than controls. TLR4 expression was significantly lower in chronic UTI patients than in acute pyelonephritis or healthy controls. CXCR1 expression was similar in both controls and patients. TLR4 expression in chronic UTI patients after astragalus treatment was higher than pre-treatment. CONCLUSIONS: The results indicate the relationship between the carrier status of TLR4 (896) G alleles and the development of UTI, especially acute cystitis and urethritis, in adults. TLR4 expression levels are correlated with chronic UTI.


Asunto(s)
Regulación de la Expresión Génica , Polimorfismo Genético , Receptor Toll-Like 4/genética , Infecciones Urinarias/genética , Infecciones Urinarias/microbiología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Planta del Astrágalo/metabolismo , Estudios de Casos y Controles , Femenino , Perfilación de la Expresión Génica , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Extractos Vegetales/farmacología , Receptores de Interleucina-8A/metabolismo
10.
Biochem Biophys Res Commun ; 397(2): 232-8, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20546703

RESUMEN

The innate host defenses at mucosal surfaces are critical in the early stages of urinary tract bacterial infection. Recent studies have shown that uroepithelial cells aid innate immune cells in fighting off infection, although the exact mechanism by which the uroepithilium participates in immunity remains unclear. TLR4 has been implicated to possess antimicrobial activities specific for bladder epithelial cells (BECs). TLR4 promotes secretion of IL-6 and IL-8, mediates inhibition of bladder epithelial cell (BEC) bacterial invasion, and mediates expulsion of uropathogenic Escherichia coli from BECs. In this study, cultured 5637 cells and Balb/C mice were treated with Astragalus polysaccharides (APS) against invading E. coli. To determine the contribution of TLR4 upregulation to immune response, TLR4 expression and bacterial colony numbers were monitored. After 24 h incubation, only 5637 cells treated with 500 microg/ml APS expressed higher levels of TLR4 compared with the untreated group. However, after 48h, all 5637 cells treated by APS showed higher levels of TLR4 expression than the control cells. The TLR4 expression in the bladder and macrophages mice that received APS was higher than that in the controls. Bacterial colonization in 5637 cells and the bladders of mice treated with APS was significantly reduced compared with the controls. These results demonstrate that at certain concentrations, APS can induce increased TLR4 expression in vivo and in vitro. Further, TLR4 expression upregulation can enhance innate immunity during mucosal bacterial infection. The findings establish the use of APS to modulate the innate immune response of the urinary tract through TLR4 expression regulation as an alternative option for UTI treatment.


Asunto(s)
Planta del Astrágalo/química , Inmunidad Innata/efectos de los fármacos , Polisacáridos/farmacología , Receptor Toll-Like 4/biosíntesis , Vejiga Urinaria/efectos de los fármacos , Infecciones Urinarias/tratamiento farmacológico , Animales , Línea Celular , Células Epiteliales/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Femenino , Humanos , Interleucina-6/biosíntesis , Interleucina-8/sangre , Ratones , Ratones Endogámicos BALB C , Polisacáridos/uso terapéutico , ARN Mensajero/biosíntesis , Regulación hacia Arriba , Vejiga Urinaria/inmunología , Infecciones Urinarias/inmunología
11.
Antiviral Res ; 84(1): 23-30, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19646486

RESUMEN

Hepatitis C virus (HCV) has emerged as the major pathogen of liver disease worldwide. The mechanisms of HCV infection and interaction with a host are poorly understood. What exactly is required for efficient control of HCV infection is largely unknown. Standard treatment combining interferon-alpha (IFN-alpha) and ribavirine is effective in about 50% of the treated patients, however associated with significant toxicity and cost. Therefore, the development of new drugs or vaccines is urgently needed. An efficient vaccine against HCV infection requires induction of broad cellular and humoral immune responses against several viral proteins. We have engineered the combined vaccine candidate mT+mE1, an inclusion of multiple epitopes from HCV NS3, core (C) and E1 proteins. mT contains multiple conserved CD4(+) and CD8(+) T cell epitopes from HCV NS3 and C proteins. mE1 is based on eight dominant neutralizing epitopes of E1 protein from six HCV genotypes. In current study, we showed that immunization with mT+mE1 induced high titers of IgG, IgG1 and IgG2a antibodies to mE1, and high level of NS3- or C-specific CTLs. Furthermore, mT+mE1 elicited a Th1-biased immune response with secretion of high amounts of IFN-gamma, compared with mT alone. Prophylactic as well as therapeutic administration of mT+mE1 in BALB/c mice led to protecting mice against SP2/0 tumor cells expressing HCV NS3 protein. These results suggested that mT+mE1 elicited strong humoral immune responses and multiple specific cellular immune responses. The vaccine candidate is now being tested in pre-clinical trials.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/inmunología , Proteínas del Núcleo Viral/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Epítopos/administración & dosificación , Epítopos/genética , Epítopos/inmunología , Femenino , Hepacivirus/genética , Hepatitis C/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Ingeniería de Proteínas , Vacunas Combinadas/administración & dosificación , Proteínas del Núcleo Viral/administración & dosificación , Proteínas del Núcleo Viral/genética , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Vacunas contra Hepatitis Viral/administración & dosificación , Vacunas contra Hepatitis Viral/genética , Proteínas no Estructurales Virales/administración & dosificación , Proteínas no Estructurales Virales/genética
12.
Microb Drug Resist ; 14(2): 145-50, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18489241

RESUMEN

The beta-lactamase (BLA) genes, the genes for aminoglycosides-modifying enzymes (AMEs), disinfectant-sulfanilamide resistance (qacEDelta1-sul1) genes, class 1 integrase (intl1) gene, and the qnr gene associated with plasmid-mediated quinolone resistance were analyzed using PCR and verified by DNA sequencing for 31 clinical isolates of multidrug-resistant Acinetobacter baumannii (MDRAB). The organism typing was performed by pulsed-field gel electrophoresis (PFGE). The positive rate of ADC, TEM, PER, and DHA of BLA genes were 100%, 61.3%, 19.4%, and 3.2%, respectively; however, the genes of SHV, OXA-23 group, OXA-24 group, GES, VIM, IMP, and qnr gene were negative. The positive rate of the genes of AMEs for aac (3)-I, aac (6')-I, ant (3")-I, ant (2")-I, aac (3)-II, and aac (6')-II were 67.7%, 45.2%, 29.0%, 22.6%, 12.9%, and 3.2%, respectively. The positive rate of qacEDelta1-sul1 and intl1 were 80.6% and 58.1%, respectively. Six different PFGE clones were found, of which two dominated. The findings show that clinical isolates of MDRAB harbor various kinds of resistance genes.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana Múltiple/genética , Infecciones por Acinetobacter/epidemiología , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/clasificación , Acinetobacter baumannii/genética , Acinetobacter baumannii/aislamiento & purificación , China/epidemiología , Infección Hospitalaria/epidemiología , Infección Hospitalaria/microbiología , Electroforesis en Gel de Campo Pulsado , Humanos , Pruebas de Sensibilidad Microbiana , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA