Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(13): 2265-2278.e14, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35568034

RESUMEN

Breakthrough infections by SARS-CoV-2 variants become the global challenge for pandemic control. Previously, we developed the protein subunit vaccine ZF2001 based on the dimeric receptor-binding domain (RBD) of prototype SARS-CoV-2. Here, we developed a chimeric RBD-dimer vaccine approach to adapt SARS-CoV-2 variants. A prototype-Beta chimeric RBD-dimer was first designed to adapt the resistant Beta variant. Compared with its homotypic forms, the chimeric vaccine elicited broader sera neutralization of variants and conferred better protection in mice. The protection of the chimeric vaccine was further verified in macaques. This approach was generalized to develop Delta-Omicron chimeric RBD-dimer to adapt the currently prevalent variants. Again, the chimeric vaccine elicited broader sera neutralization of SARS-CoV-2 variants and conferred better protection against challenge by either Delta or Omicron SARS-CoV-2 in mice. The chimeric approach is applicable for rapid updating of immunogens, and our data supported the use of variant-adapted multivalent vaccine against circulating and emerging variants.


Asunto(s)
COVID-19 , Vacunas , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Ratones , SARS-CoV-2/genética
2.
Cell ; 185(10): 1728-1744.e16, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35460644

RESUMEN

As the emerging variants of SARS-CoV-2 continue to drive the worldwide pandemic, there is a constant demand for vaccines that offer more effective and broad-spectrum protection. Here, we report a circular RNA (circRNA) vaccine that elicited potent neutralizing antibodies and T cell responses by expressing the trimeric RBD of the spike protein, providing robust protection against SARS-CoV-2 in both mice and rhesus macaques. Notably, the circRNA vaccine enabled higher and more durable antigen production than the 1mΨ-modified mRNA vaccine and elicited a higher proportion of neutralizing antibodies and distinct Th1-skewed immune responses. Importantly, we found that the circRNARBD-Omicron vaccine induced effective neutralizing antibodies against the Omicron but not the Delta variant. In contrast, the circRNARBD-Delta vaccine protected against both Delta and Omicron or functioned as a booster after two doses of either native- or Delta-specific vaccination, making it a favorable choice against the current variants of concern (VOCs) of SARS-CoV-2.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Macaca mulatta , Ratones , ARN Circular/genética , SARS-CoV-2/genética , Vacunas Sintéticas/genética , Vacunas de ARNm
3.
Nature ; 586(7830): 572-577, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32726802

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a respiratory disease called coronavirus disease 2019 (COVID-19), the spread of which has led to a pandemic. An effective preventive vaccine against this virus is urgently needed. As an essential step during infection, SARS-CoV-2 uses the receptor-binding domain (RBD) of the spike protein to engage with the receptor angiotensin-converting enzyme 2 (ACE2) on host cells1,2. Here we show that a recombinant vaccine that comprises residues 319-545 of the RBD of the spike protein induces a potent functional antibody response in immunized mice, rabbits and non-human primates (Macaca mulatta) as early as 7 or 14 days after the injection of a single vaccine dose. The sera from the immunized animals blocked the binding of the RBD to ACE2, which is expressed on the cell surface, and neutralized infection with a SARS-CoV-2 pseudovirus and live SARS-CoV-2 in vitro. Notably, vaccination also provided protection in non-human primates to an in vivo challenge with SARS-CoV-2. We found increased levels of RBD-specific antibodies in the sera of patients with COVID-19. We show that several immune pathways and CD4 T lymphocytes are involved in the induction of the vaccine antibody response. Our findings highlight the importance of the RBD domain in the design of SARS-CoV-2 vaccines and provide a rationale for the development of a protective vaccine through the induction of antibodies against the RBD domain.


Asunto(s)
Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , COVID-19 , Vacunas contra la COVID-19 , Humanos , Macaca mulatta/inmunología , Macaca mulatta/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Modelos Moleculares , Dominios Proteicos , SARS-CoV-2 , Suero/inmunología , Bazo/citología , Bazo/inmunología , Linfocitos T/inmunología , Vacunación
4.
FASEB J ; 38(5): e23500, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38441537

RESUMEN

Hepatitis E virus (HEV) persists in the male genital tract that associates with infertility. However, the presence of HEV in the female genital tract is unreported. Vaginal secretions, cervical smears, and cervix uteri were collected to explore the presence of HEV in the female genital tract. HEV RNA and/or antigens were detected in the vaginal secretions, cervical smears, and the cervix uteri of women. The infectivity of HEV excreted into vaginal secretions was further validated in vitro. In addition, HEV replicates in the female genital tract were identified in HEV-infected animal models by vaginal injection or vaginal mucosal infection to imitate sexual transmission. Serious genital tract damage and inflammatory responses with significantly elevated mucosal innate immunity were observed in women or animals with HEV vaginal infection. Results demonstrated HEV replicates in the female genital tract and causes serious histopathological damage and inflammatory responses.


Asunto(s)
Líquidos Corporales , Hepatitis A , Virus de la Hepatitis E , Hepatitis E , Animales , Femenino , Masculino , Humanos , Vagina
5.
Artículo en Inglés | MEDLINE | ID: mdl-38877781

RESUMEN

Hepatitis E virus (HEV) is the major pathogen of viral hepatitis. Immunocompromised individuals infected by HEV are prone to chronic hepatitis and increase the risk of hepato-cellular carcinoma (HCC). Inhibitor of growth family member 5 (ING5) is a tumor suppressor that is expressed at low levels in cancer tumors or cells. However, the underlying relationship between ING5 and HEV infection is unclear. In the present study, acute and chronic HEV animal models are used to explore the interaction between ING5 and HEV. Notably, the expression of ING5 is significantly increased in both the livers of acute HEV-infected BALB/c mice and chronic HEV-infected rhesus macaques. In addition, the relationship between HEV infection and ING5 expression is further identified in human hepatoma (HepG-2) cells. In conclusion, HEV infection strongly upregulates ING5 expression both in vivo and in vitro, which has significant implications for further understanding the pathogenic mechanism of HEV infection.

6.
J Med Virol ; 95(6): e28846, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37282766

RESUMEN

Since the first SARS-CoV-2 outbreak in late 2019, the SARS-CoV-2 genome has harbored multiple mutations, especially spike protein mutations. The currently fast-spreading Omicron variant that manifests without symptoms or with upper respiratory diseases has been recognized as a serious global public health problem. However, its pathological mechanism is largely unknown. In this work, rhesus macaques, hamsters, and BALB/C mice were employed as animal models to explore the pathogenesis of Omicron (B.1.1.529). Notably, Omicron (B.1.1.529) infected the nasal turbinates, tracheae, bronchi, and lungs of hamsters and BALB/C mice with higher viral loads than in those of rhesus macaques. Severe histopathological damage and inflammatory responses were observed in the lungs of Omicron (B.1.1.529)-infected animals. In addition, viral replication was found in multiple extrapulmonary organs. Results indicated that hamsters and BALB/c mice are potential animal models for studies on the development of drugs/vaccines and therapies for Omicron (B.1.1.529).


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Animales , Cricetinae , Macaca mulatta , Ratones Endogámicos BALB C , Bronquios
7.
FASEB J ; 36(1): e22104, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34918388

RESUMEN

MicroRNAs (miRNAs), the non-coding RNAs of ~22 nucleotides (nt) in length, play a vital role in regulating viral replication. Hepatitis E virus (HEV), a single-stranded RNA virus, is a predominant pathogen of acute hepatitis worldwide. Virus-encoded miRNAs regulate the viral life cycle and escape from the host innate immune system. However, it is rarely known about HEV-encoded miRNA (HEV-miR-A6). In the present study, HEV-miR-A6 was screened by microarray, and further identified in vivo and in vitro. HEV-miR-A6 originated from the methylase (MeT) of HEV open reading frame 1 (ORF1) and was highly conserved in eight HEV genotypes. HEV-miR-A6 expression was growing during HEV replication, and significantly increased in acute hepatitis E patients than convalescence patients. Furthermore, HEV-miR-A6 was specifically detected in liver, spleen, kidney and colon by in situ hybridization. To identify the specificity of HEV-miR-A6, its mutants (HEV-miR-A6M1 and HEV-miR-A6M2) were constructed to change the stem-loop structure. Interestingly, over-expression of HEV-miR-A6 or HEV-miR-A6M1 significantly facilitated viral replication, while HEV-miR-A6M2, another mutant completely changed the stem-loop structure was invalid. SIRP-α, a candidate target gene of HEV-miR-A6, was activated when HEV-miR-A6 over-expressed to inhibit the phosphorylation of IRF3, and subsequently suppressed the expression of type I interferon ß (IFN-ß). The promotion of viral replication by HEV-miR-A6 further identified in vivo. Significant suppression of IFN-ß production in the serum of HEV-infected mice pre-treated with HEV-miR-A6 was observed. In summary, HEV-miR-A6 activates SIRP-α to promote viral replication by inhibition of IFN-ß expression.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis E/fisiología , Hepatitis E/metabolismo , Interferón beta/metabolismo , MicroARNs/biosíntesis , ARN Viral/biosíntesis , Replicación Viral , Femenino , Humanos , Masculino , Especificidad de Órganos
8.
Virol J ; 20(1): 111, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37264422

RESUMEN

BACKGROUND: Hepatitis E virus (HEV) infection is a common cause of acute hepatitis worldwide and causes approximately 30% case fatality rate among pregnant women. Pregnancy serum (PS), which contains a high concentration of estradiol, facilitates HEV replication in vitro through the suppression of the PI3K-AKT-mTOR and cAMPK-PKA-CREB signaling pathways. However, the proteomics of the complex host responses to HEV infection, especially how PS facilitates viral replication, remains unclear. METHODS: In this study, the differences in the proteomics of HEV-infected HepG2 cells supplemented with fetal bovine serum (FBS) from those of HEV-infected HepG2 cells supplemented with serum from women in their third trimester of pregnancy were quantified by using isobaric tags for relative and absolute quantification technology. RESULTS: A total of 1511 proteins were identified, among which 548 were defined as differentially expressed proteins (DEPs). HEV-infected cells supplemented with PS exhibited the most significant changes at the protein level. A total of 328 DEPs, including 66 up-regulated and 262 down-regulated proteins, were identified in HEV-infected cells supplemented with FBS, whereas 264 DEPs, including 201 up-regulated and 63 down-regulated proteins, were found in HEV-infected cells supplemented with PS. Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that in HEV-infected cells, PS supplementation adjusted more host genes and signaling pathways than FBS supplementation. The DEPs involved in virus-host interaction participated in complex interactions, especially a large number of immune-related protein emerged in HEV-infected cells supplemented with PS. Three significant or interesting proteins, including filamin-A, thioredoxin, and cytochrome c, in HEV-infected cells were functionally verified. CONCLUSIONS: The results of this study provide new and comprehensive insight for exploring virus-host interactions and will benefit future studies on the pathogenesis of HEV in pregnant women.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Femenino , Humanos , Embarazo , Virus de la Hepatitis E/genética , Proteómica/métodos , Fosfatidilinositol 3-Quinasas/genética , Genotipo , Replicación Viral
9.
Liver Int ; 43(2): 317-328, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36305303

RESUMEN

BACKGROUND: Hepatitis E virus (HEV) infection causes serious adverse pregnancy outcomes during pregnancy. However, the maternal and fetal damage induced by HEV infection is rarely reported. METHODS: A BALB/c pregnant mouse model was established to explore the maternal and fetal pathological damage and inflammatory responses caused by HEV infection. RESULTS: Notably, miscarriages and stillbirths were observed in HEV-infected pregnant mice. HEV infections were identified by qRT-PCR, immunohistochemical analysis and immunofluorescence assay in the uterus, placenta, umbilical cords and livers and brains of fetuses. Serious inflammatory responses and pathological damage were triggered in the uterus and placenta of HEV-infected pregnant mice. Vertical transmission of HEV resulted in severe pathological damage and inflammatory responses in the livers and brains of fetuses, as well as emerging apoptosis cells in the brains of fetuses. Most of the cytokines/chemokines in the sera were significantly increased in the HEV-infected pregnant mice. Remarkably, cytokines/chemokines were significantly different between HEV-infected pregnant and miscarriage mice; IL9, GM-CSF and IL1α were the most important three cytokines/chemokines in determining the pregnancy outcomes. CONCLUSION: HEV infections cause serious maternal/fetal pathological damage, inflammatory responses and apoptosis, which may be responsible for adverse pregnancy outcomes.


Asunto(s)
Aborto Espontáneo , Virus de la Hepatitis E , Hepatitis E , Complicaciones Infecciosas del Embarazo , Animales , Femenino , Ratones , Embarazo , Aborto Espontáneo/etiología , Citocinas , Hepatitis E/complicaciones , Hepatitis E/patología , Ratones Endogámicos BALB C
10.
Gastroenterology ; 160(5): 1647-1661, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33307034

RESUMEN

BACKGROUND & AIMS: Gastrointestinal (GI) manifestations have been increasingly reported in patients with coronavirus disease 2019 (COVID-19). However, the roles of the GI tract in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are not fully understood. We investigated how the GI tract is involved in SARS-CoV-2 infection to elucidate the pathogenesis of COVID-19. METHODS: Our previously established nonhuman primate (NHP) model of COVID-19 was modified in this study to test our hypothesis. Rhesus monkeys were infected with an intragastric or intranasal challenge with SARS-CoV-2. Clinical signs were recorded after infection. Viral genomic RNA was quantified by quantitative reverse transcription polymerase chain reaction. Host responses to SARS-CoV-2 infection were evaluated by examining inflammatory cytokines, macrophages, histopathology, and mucin barrier integrity. RESULTS: Intranasal inoculation with SARS-CoV-2 led to infections and pathologic changes not only in respiratory tissues but also in digestive tissues. Expectedly, intragastric inoculation with SARS-CoV-2 resulted in the productive infection of digestive tissues and inflammation in both the lung and digestive tissues. Inflammatory cytokines were induced by both types of inoculation with SARS-CoV-2, consistent with the increased expression of CD68. Immunohistochemistry and Alcian blue/periodic acid-Schiff staining showed decreased Ki67, increased cleaved caspase 3, and decreased numbers of mucin-containing goblet cells, suggesting that the inflammation induced by these 2 types of inoculation with SARS-CoV-2 impaired the GI barrier and caused severe infections. CONCLUSIONS: Both intranasal and intragastric inoculation with SARS-CoV-2 caused pneumonia and GI dysfunction in our rhesus monkey model. Inflammatory cytokines are possible connections for the pathogenesis of SARS-CoV-2 between the respiratory and digestive systems.


Asunto(s)
COVID-19/transmisión , Gastroenteritis/patología , Tracto Gastrointestinal/patología , Pulmón/patología , Animales , Bronquios/metabolismo , Bronquios/patología , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/patología , Prueba de Ácido Nucleico para COVID-19 , Caspasa 3/metabolismo , Citocinas/inmunología , Modelos Animales de Enfermedad , Mucosa Gástrica , Gastroenteritis/metabolismo , Gastroenteritis/virología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Células Caliciformes/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Antígeno Ki-67/metabolismo , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/metabolismo , Macaca mulatta , Mucosa Nasal , ARN Viral/aislamiento & purificación , Distribución Aleatoria , Recto/metabolismo , Recto/patología , SARS-CoV-2 , Tráquea/metabolismo , Tráquea/patología
12.
J Cell Mol Med ; 25(7): 3361-3370, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33682317

RESUMEN

In this study, the purpose of this study was to investigate the role of TNFAIP8 in gastric cancer (GC). The expression of TNFAIP8 was detected by RT-PCR or western blot . TNFAIP8 was silenced or overexpressed in two cell lines. CCK-8 assay, transwell assay and flow cytometry were used to analyse cell viability, cell invasion capability and apoptosis, respectively. Nude mice were inoculated with TNFAIP8 silencing or overexpressing cells to form transplanted tumours. HE staining and immunohistochemistry assay were performed to assess histopathological changes in tumours. We found that the mRNA and protein expression of TNFAIP8 were significantly up-regulated in GC tumour tissues and cells compared with the normal counterparts. Overexpression of TNFAIP8 in GC cells increased cell viability, decreased apoptosis and promoted the cell migration ability. Meanwhile, increased expression of TNFAIP8 promoted autophagy, while inhibiting mTOR-Akt-ULK1 signal pathway. In conclusions, this study presents data that TNFAIP8 inhibits GC cells presumably by down-regulating mTOR-Akt-ULK1 signal pathway and activating autophagy signal.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Transducción de Señal , Neoplasias Gástricas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba
13.
J Viral Hepat ; 28(8): 1219-1229, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33894039

RESUMEN

Hepatitis E virus (HEV) infection is the most common cause of acute viral hepatitis worldwide. However, host-HEV interactions have yet to be fully understood. Zinc-finger antiviral protein (ZAP) is a novel interferon (IFN)-stimulated gene product that inhibits a variety of viruses in synergy with IFN-ß. To evaluate the role of ZAP in HEV infection, its expressions in HEV-infected patients and in cell cultures were measured. We report a significant inhibition of ZAP expression in patients with HEV genotype four acute infection. The expression of ZAP in the HEV life cycle was monitored in cultures of HEV-infected cells. Results indicated that the ZAP level decreased significantly after HEV infection. ZAP over-expression inhibited HEV replication, whereas its knockdown by RNA interference significantly increased HEV RNA. These suggest that ZAP serves as an antiviral in HEV infection. Moreover, silencing ZAP decreased IFN regulatory factor 3 (IRF3) phosphorylation in HEV-infected cells treated with poly(I:C), indicating that ZAP synergizes with IFN-ß. In conclusion, ZAP is an important anti-HEV host factor and in synergy with IFN-ß, inhibits HEV replication.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Antivirales/farmacología , Hepatitis E/tratamiento farmacológico , Humanos , Replicación Viral , Zinc
14.
J Med Virol ; 93(11): 6267-6277, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34076903

RESUMEN

Hepatitis E virus (HEV) is the major pathogen of viral hepatitis. However, the understanding of the HEV life cycle is limited. In the present study, cells were separately infected with nonenveloped HEV (derived from feces or bile) or quasi-enveloped HEV (derived from the cell culture after serial passages, eHEV) and observed by confocal fluorescence microscopy to investigate the life cycle of HEV. HEV finished its binding and entry into host cells at first 6 h postinoculation (hpi). Cells inoculated with eHEV showed less infectivity than cells inoculated with nonenveloped HEV. Newly synthesized progeny virions were released into the supernatant of cell cultures from 48 hpi. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis results showed that the supernatant's progeny viruses were infectious even after five serial passages. These results show the significant difference between nonenveloped HEV and eHEV, which will provide novel insights into the HEV replication cycle. The efficient cell culture of HEV will promote the development of anti-HEV drugs and vaccines.


Asunto(s)
Virus de la Hepatitis E/fisiología , Replicación Viral , Células A549 , Carcinoma Hepatocelular , Línea Celular , Línea Celular Tumoral , Hepatitis E/virología , Virus de la Hepatitis E/clasificación , Humanos , Neoplasias Hepáticas , Microscopía Fluorescente/métodos , Envoltura Viral , Virión/fisiología
15.
J Med Virol ; 93(6): 3769-3778, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33128390

RESUMEN

Hepatitis E virus (HEV) infection has become a global concern with high mortality rates among pregnant women, especially those in their third trimester of pregnancy. Estrogen plays an important role in mediating the body, regulating physiological and pathological processes. Estrogen is activated by binding to estrogen receptors (ERs) and mediates rapid signaling events by pathways that involve transmembrane ERs. Our previous study had confirmed that high estrogen levels during pregnancy are associated with high HEV titers. However, the association between HEV infection and estrogen signaling pathways remains unclear. In the present study, the regulation of estrogen signaling pathways by HEV infection was evaluated. Results demonstrated that HEV infection significantly inhibits the cAMP-PKA-CREB and PI3K-AKT-mTOR signaling pathways, but is independent of the Ras-Raf-MEK-ERK signaling pathway. In summary, the increasing estrogen levels and highly activated ERα during pregnancy aggravates HEV replication. The exacerbation of HEV replication, in turn, inhibits ERα expression and suppresses both cAMP-PKA-CREB and PI3K-AKT-mTOR signaling pathways.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Estrógenos/metabolismo , Virus de la Hepatitis E/patogenicidad , Hepatitis E/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Células A549 , Estrógenos/genética , Femenino , Humanos , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Med Sci Monit ; 27: e929879, 2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34531359

RESUMEN

BACKGROUND Hepatocellular carcinoma (HCC) is the second leading cause of cancer deaths. Transarterial chemoembolization (TACE) has been widely applied for treating patients with unresectable HCC. This study explored the factors influencing early recurrence (ER) after TACE in HCC patients. MATERIAL AND METHODS A total of 197 patients were divided into the ER group and the non-ER group. Univariate and multivariate Cox regression analyses were carried out to explore the influencing factors. Univariate Kaplan-Meier survival curves and restricted cubic splines were plotted for visualizing the relations between the influencing factors and ER. RESULTS According to the multivariate analysis, for every 1-cm increase in the maximum tumor diameter, the risk of ER increased by 0.235 times (95% CI: 1.144-1.333, P<0.001). Patients with adjacent lobe invasion had a 1.227-fold higher risk of ER than those without (95% CI: 1.461-3.394, P<0.001). For every unit increase in neutrophil-to-lymphocyte ratio (NLR), the risk increased by 0.107-fold (95% CI: 1.012-1.211, P=0.027). Compared to patients at the very early/early Barcelona clinic liver cancer (BCLC) stage, those at the advanced/end stage had a 2.045-fold increased risk of ER (95% CI: 1.259-7.366, P=0.014). CONCLUSIONS The maximum tumor diameter, adjacent lobe invasion, NLR, and advanced/end stage BCLC stage were all risk factors for ER after TACE in HCC patients.


Asunto(s)
Carcinoma Hepatocelular/epidemiología , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica/métodos , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/terapia , Recurrencia Local de Neoplasia/epidemiología , Adulto , Anciano , Anciano de 80 o más Años , China/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
17.
J Immunol ; 201(9): 2557-2569, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30282753

RESUMEN

Enterovirus D68 (EV-D68) belongs to the picornavirus family and was first isolated in CA, USA, in 1962. EV-D68 can cause severe cranial nerve system damage such as flaccid paralysis and acute respiratory diseases such as pneumonia. There are currently no efficient therapeutic methods or effective prophylactics. In this study, we isolated the mAb A6-1 from an EV-D68-infected rhesus macaque (Macaca mulatta) and found that the Ab provided effective protection in EV-D68 intranasally infected suckling mice. We observed that A6-1 bound to the DE loop of EV-D68 VP1 and interfered with the interaction between the EV-D68 virus and α2,6-linked sialic acids of the host cell. The production of A6-1 and its Ab properties present a bridging study for EV-D68 vaccine design and provide a tool for analyzing the process by which Abs can inhibit EV-D68 infection.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas de la Cápside/inmunología , Infecciones por Enterovirus/prevención & control , Enterovirus/inmunología , Vacunas Virales/inmunología , Secuencia de Aminoácidos/genética , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Proteínas de la Cápside/genética , Enterovirus Humano D , Infecciones por Enterovirus/inmunología , Femenino , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Ácidos Siálicos/metabolismo , Acoplamiento Viral
18.
J Gastroenterol Hepatol ; 35(9): 1549-1554, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32103514

RESUMEN

BACKGROUND AND AIM: Following abdominal surgery, patients usually experience a transient episode of impaired gastrointestinal motility. This study aimed to determine whether a single preoperative dose of dexamethasone can promote the recovery of gastrointestinal function in patients following elective gastrointestinal surgery. METHODS: In this single-center, two-arm, parallel, randomized controlled trial, we studied 126 patients (aged 18-80 years) who underwent elective open or laparoscopic bowel surgery for malignant or benign pathology. At the induction of anesthesia, a treatment group (n = 64) received a single dose of 8-mg intravenous dexamethasone, and a control group (n = 62) received normal saline. RESULTS: Intravenous administration of 8-mg dexamethasone significantly decreased the time to return of flatus by an average of approximately 8 h (P < 0.05). Abdominal distension was significantly reduced on the third day after surgery in the dexamethasone group (P < 0.05), and the time to tolerance of a liquid diet was shorter in the dexamethasone group (P < 0.01). There were no significant differences in other secondary outcomes, including postoperative pain, complication rates, length of hospital stay, or time to first defecation, between the two groups. CONCLUSIONS: A single intravenous dose of 8-mg dexamethasone at induction of anesthesia significantly decreases the time to return of flatus, improves abdominal distension at 72 h, and promotes tolerance of a liquid diet. Although further studies are required to confirm our results, we recommend that dexamethasone should be used more widely in gastrointestinal surgery.


Asunto(s)
Dexametasona/administración & dosificación , Enfermedades Gastrointestinales/prevención & control , Enfermedades Gastrointestinales/fisiopatología , Motilidad Gastrointestinal , Complicaciones Posoperatorias/prevención & control , Cuidados Preoperatorios , Recuperación de la Función , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Endoscopía Gastrointestinal/métodos , Humanos , Infusiones Intravenosas , Laparoscopía/métodos , Persona de Mediana Edad , Complicaciones Posoperatorias/fisiopatología , Resultado del Tratamiento , Adulto Joven
19.
BMC Infect Dis ; 18(1): 687, 2018 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-30572833

RESUMEN

BACKGROUND: Hepatitis E virus (HEV) is a leading cause of hepatitis worldwide. However, its infection biology and pathogenesis remain largely elusive. Furthermore, no proven medication is available for treating hepatitis E. Robust experimental models are urgently required to advance the research of HEV infection. Because of the lacking of a sophisticated small animal model, this study aimed to establish a mouse model of HEV infection. METHODS: We constructed a full-length swine HEV cDNA clone of genotype 4 (named as pGEM-HEV) by reverse genetics approach. And we inoculated with HEV RNA in BALB/c mice to establish small animal model for HEV infection and pathogenesis studies. RESULTS: The capped RNA transcripts of pGEM-HEV prepared in vitro were replication-competent in HepG2 cells. Importantly, BALB/c mice intravenously inoculated with RNA transcripts of pGEM-HEV developed an active infection as shown by shedding viruses in feces, detectable negative strand of HEV in the liver, spleen and kidney, and causing liver inflammation. CONCLUSION: In this study, we successfully established of BALB/c mice-based small animal model for HEV provides an opportunity to further understand HEV pathogenesis and to develop effective antiviral medications.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Hepatitis E/genética , Hepatitis E/virología , Ratones Endogámicos BALB C , Genética Inversa/métodos , Porcinos/virología , Animales , Línea Celular Tumoral , Clonación de Organismos/métodos , Femenino , Células Hep G2 , Hepatitis E/genética , Hepatitis E/patología , Virus de la Hepatitis E/patogenicidad , Humanos , Ratones , ARN Viral/genética , Enfermedades de los Porcinos/patología , Enfermedades de los Porcinos/virología , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA