Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(4): e0006424, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38488360

RESUMEN

As one of the most important causative agents of severe gastroenteritis in children, piglets, and other young animals, species A rotaviruses have adversely impacted both human health and the global swine industry. Vaccines against rotaviruses (RVs) are insufficiently effective, and no specific treatment is available. To understand the relationships between porcine RV (PoRV) infection and enterocytes in terms of the cellular lipid metabolism, we performed an untargeted liquid chromatography mass spectrometry (LC-MS) lipidomics analysis of PoRV-infected IPEC-J2 cells. Herein, a total of 451 lipids (263 upregulated lipids and 188 downregulated lipids), spanning sphingolipid, glycerolipid, and glycerophospholipids, were significantly altered compared with the mock-infected group. Interestingly, almost all the ceramides among these lipids were upregulated during PoRV infection. LC-MS analysis was used to validated the lipidomics data and demonstrated that PoRV replication increased the levels of long-chain ceramides (C16-ceramide, C18-ceramide, and C24-ceramide) in cells. Furthermore, we found that these long-chain ceramides markedly inhibited PoRV infection and that their antiviral actions were exerted in the replication stage of PoRV infection. Moreover, downregulation of endogenous ceramides with the ceramide metabolic inhibitors enhanced PoRV propagation. Increasing the levels of ceramides by the addition of C6-ceramide strikingly suppressed the replication of diverse RV strains. We further found that the treatment with an apoptotic inhibitor could reverse the antiviral activity of ceramide against PoRV replication, demonstrating that ceramide restricted RV infection by inducing apoptosis. Altogether, this study revealed that ceramides played an antiviral role against RV infection, providing potential approaches for the development of antiviral therapies.IMPORTANCERotaviruses (RVs) are among the most important zoonosis viruses, which mainly infected enterocytes of the intestinal epithelium causing diarrhea in children and the young of many mammalian and avian species. Lipids play an essential role in viral infection. A comprehensive understanding of the interaction between RV and lipid metabolism in the enterocytes will be helpful to control RV infection. Here, we mapped changes in enterocyte lipids following porcine RV (PoRV) infection using an untargeted lipidomics approach. We found that PoRV infection altered the metabolism of various lipid species, especially ceramides (derivatives of the sphingosine). We further demonstrated that PoRV infection increased the accumulation of ceramides and that ceramides exerted antiviral effects on RV replication by inducing apoptosis. Our findings fill a gap in understanding the alterations of lipid metabolism in RV-infected enterocytes and highlight the antiviral effects of ceramides on RV infection, suggesting potential approaches to control RV infection.


Asunto(s)
Ceramidas , Infecciones por Rotavirus , Rotavirus , Animales , Ceramidas/metabolismo , Metabolismo de los Lípidos , Lipidómica , Rotavirus/fisiología , Porcinos , Enterocitos/metabolismo , Enterocitos/virología , Infecciones por Rotavirus/metabolismo , Línea Celular
2.
PLoS Pathog ; 15(10): e1008057, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31671153

RESUMEN

Human astroviruses (HAstV) are understudied positive-strand RNA viruses that cause gastroenteritis mostly in children and the elderly. Three clades of astroviruses, classic, MLB-type and VA-type have been reported in humans. One limitation towards a better understanding of these viruses has been the lack of a physiologically relevant cell culture model that supports growth of all clades of HAstV. Herein, we demonstrate infection of HAstV strains belonging to all three clades in epithelium-only human intestinal enteroids (HIE) isolated from biopsy-derived intestinal crypts. A detailed investigation of infection of VA1, a member of the non-canonical HAstV-VA/HMO clade, showed robust replication in HIE derived from different patients and from different intestinal regions independent of the cellular differentiation status. Flow cytometry and immunofluorescence analysis revealed that VA1 infects several cell types, including intestinal progenitor cells and mature enterocytes, in HIE cultures. RNA profiling of VA1-infected HIE uncovered that the host response to infection is dominated by interferon (IFN)-mediated innate immune responses. A comparison of the antiviral host response in non-transformed HIE and transformed human colon carcinoma Caco-2 cells highlighted significant differences between these cells, including an increased magnitude of the response in HIE. Additional studies confirmed the sensitivity of VA1 to exogenous IFNs, and indicated that the endogenous IFN response of HIE to curtail the growth of strains from all three clades. Genotypic variation in the permissiveness of different HIE lines to HAstV could be overcome by pharmacologic inhibition of JAK/STAT signaling. Collectively, our data identify HIE as a universal infection model for HAstV and an improved model of the intestinal epithelium to investigate enteric virus-host interactions.


Asunto(s)
Infecciones por Astroviridae/inmunología , Infecciones por Astroviridae/veterinaria , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Mamastrovirus/fisiología , Tropismo Viral/genética , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Enterocitos/virología , Gastroenteritis/virología , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/virología , Intestino Delgado/citología , Intestino Delgado/virología , Mamastrovirus/genética , Mamastrovirus/inmunología , Células Vero , Tropismo Viral/inmunología
3.
RNA Biol ; 18(5): 796-808, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33406999

RESUMEN

The pathogenic human enterovirus EV-A71 has raised serious public health concerns. A hallmark of EV-A71 infection is the distortion of host transcriptomes in favour of viral replication. While high-throughput approaches have been exploited to dissect these gene dysregulations, they do not fully capture molecular perturbations at the single-cell level and in a physiologically relevant context. In this study, we applied a single-cell RNA sequencing approach on infected differentiated enterocyte cells (C2BBe1), which model the gastrointestinal epithelium targeted initially by EV-A71. Our single-cell analysis of EV-A71-infected culture provided several lines of illuminating observations: 1) This systems approach demonstrated extensive cell-to-cell variation in a single culture upon viral infection and delineated transcriptomic differences between the EV-A71-infected and bystander cells. 2) By analysing expression profiles of known EV-A71 receptors and entry facilitation factors, we found that ANXA2 was closely correlated in expression with the viral RNA in the infected population, supporting its role in EV-A71 entry in the enteric cells. 3) We further catalogued dysregulated lncRNAs elicited by EV-A71 infection and demonstrated the functional implication of lncRNA CYTOR in promoting EV-A71 replication. Viewed together, our single-cell transcriptomic analysis illustrated at the single-cell resolution the heterogeneity of host susceptibility to EV-A71 and revealed the involvement of lncRNAs in host antiviral response.


Asunto(s)
Enterovirus Humano A/patogenicidad , Interacciones Huésped-Patógeno/genética , Transcriptoma , Células Cultivadas , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/virología , Enterovirus Humano A/genética , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/virología , ARN Largo no Codificante/genética , Análisis de la Célula Individual , Replicación Viral/genética
4.
Mol Biol Rep ; 48(5): 4667-4675, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34023987

RESUMEN

The transmembrane protease serine 2 (TMPRSS2) is a membrane anchored protease that primarily expressed by epithelial cells of respiratory and gastrointestinal systems and has been linked to multiple pathological processes in humans including tumor growth, metastasis and viral infections. Recent studies have shown that TMPRSS2 expressed on cell surface of host cells could play a crucial role in activation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein which facilitates the rapid early entry of the virus into host cells. In addition, direct suppression of TMPRSS2 using small drug inhibitors has been demonstrated to be effective in decreasing SARS-CoV-2 infection in vitro, which presents TMPRSS2 protease as a potential therapeutic strategy for SARS-CoV-2 infection. Recently, SARS-CoV-2 has been shown to be capable of infecting gastrointestinal enterocytes and to provoke gastrointestinal disorders in patients with COVID-19 disease, which is considered as a new transmission route and target organ of SARS-CoV-2. In this review, we highlight the biochemical properties of TMPRSS2 protease and discuss the potential targeting of TMPRSS2 by inhibitors to prevent the SARS-CoV-2 spreading through gastro-intestinal tract system as well as the hurdles that need to be overcome.


Asunto(s)
COVID-19/metabolismo , Enterocitos/efectos de los fármacos , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Enterocitos/metabolismo , Enterocitos/virología , Humanos , SARS-CoV-2/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
5.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360898

RESUMEN

Previous studies have shown that microRNAs (miRNAs) are closely related to many viral infections. However, the molecular mechanism of how miRNAs regulate porcine epidemic diarrhea virus (PEDV) infection remains unclear. In this study, we first constructed a PEDV-infected IPEC-J2 cytopathic model to validate the relationship between miR-129a-3p expression levels and PEDV resistance. Secondly, we explored the effect of miR-129a-3p on PEDV infection by targeting the 3'UTR region of the ligand ectodysplasin (EDA) gene. Finally, transcriptome sequencing was used to analyze the downstream regulatory mechanism of EDA. The results showed that after 48 h of PEDV infection, IPEC-J2 cells showed obvious pathological changes, and miR-129a-3p expression was significantly downregulated (p < 0.01). Overexpression of miR-129a-3p mimics inhibited PEDV replication in IPEC-J2 cells; silencing endogenous miR-129a-3p can promote viral replication. A dual luciferase assay showed that miR-129a-3p could bind to the 3'UTR region of the EDA gene, which significantly reduced the expression level of EDA (p < 0.01). Functional verification showed that upregulation of EDA gene expression significantly promoted PEDV replication in IPEC-J2 cells. Overexpression of miR-129a-3p can activate the caspase activation and recruitment domain 11 (CARD11) mediated NF-κB pathway, thus inhibiting PEDV replication. The above results suggest that miR-129a-3p inhibits PEDV replication in IPEC-J2 cells by activating the NF-κB pathway by binding to the EDA 3'UTR region. Our results have laid the foundation for in-depth study of the mechanism of miR-129a-3p resistance and its application in porcine epidemic diarrhea disease-resistance breeding.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Ectodisplasinas/metabolismo , Enterocitos/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Transducción de Señal/genética , Replicación Viral/genética , Regiones no Traducidas 3' , Animales , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Regulación hacia Abajo/genética , Ectodisplasinas/genética , Enterocitos/virología , Células HEK293 , Humanos , MicroARNs/genética , Porcinos , Transfección , Regulación hacia Arriba/genética , Células Vero , Secuenciación del Exoma/métodos
6.
Rev Esp Enferm Dig ; 112(5): 383-388, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32343593

RESUMEN

Although SARS-CoV-2 may primarily enter the cells of the lungs, the small bowel may also be an important entry or interaction site, as the enterocytes are rich in angiotensin converting enzyme (ACE)-2 receptors. The initial gastrointestinal symptoms that appear early during the course of Covid-19 support this hypothesis. Furthermore, SARS-CoV virions are preferentially released apically and not at the basement of the airway cells. Thus, in the setting of a productive infection of conducting airway epithelia, the apically released SARS-CoV may be removed by mucociliary clearance and gain access to the GI tract via a luminal exposure. In addition, post-mortem studies of mice infected by SARS-CoV have demonstrated diffuse damage to the GI tract, with the small bowel showing signs of enterocyte desquamation, edema, small vessel dilation and lymphocyte infiltration, as well as mesenteric nodes with severe hemorrhage and necrosis. Finally, the small bowel is rich in furin, a serine protease which can separate the S-spike of the coronavirus into two "pinchers" (S1 and 2). The separation of the S-spike into S1 and S2 is essential for the attachment of the virion to both the ACE receptor and the cell membrane. In this special review, we describe the interaction of SARS-CoV-2 with the cell and enterocyte and its potential clinical implications.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/metabolismo , Enterocitos/virología , Enfermedades Gastrointestinales/virología , Intestino Delgado/virología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/metabolismo , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/metabolismo , COVID-19 , Infecciones por Coronavirus/virología , Enterocitos/metabolismo , Enfermedades Gastrointestinales/metabolismo , Humanos , Intestino Delgado/citología , Intestino Delgado/metabolismo , Pandemias , Neumonía Viral/virología , Receptores de Angiotensina/metabolismo , Mucosa Respiratoria/fisiología , Mucosa Respiratoria/virología , SARS-CoV-2
7.
PLoS Pathog ; 13(12): e1006768, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29211815

RESUMEN

Enteric viruses encounter diverse environments as they migrate through the gastrointestinal tract to infect their hosts. The interaction of eukaryotic viruses with members of the host microbiota can greatly impact various aspects of virus biology, including the efficiency with which viruses can infect their hosts. Mammalian orthoreovirus, a human enteric virus that infects most humans during childhood, is negatively affected by antibiotic treatment prior to infection. However, it is not known how components of the host microbiota affect reovirus infectivity. In this study, we show that reovirus virions directly interact with Gram positive and Gram negative bacteria. Reovirus interaction with bacterial cells conveys enhanced virion thermostability that translates into enhanced attachment and infection of cells following an environmental insult. Enhanced virion thermostability was also conveyed by bacterial envelope components lipopolysaccharide (LPS) and peptidoglycan (PG). Lipoteichoic acid and N-acetylglucosamine-containing polysaccharides enhanced virion stability in a serotype-dependent manner. LPS and PG also enhanced the thermostability of an intermediate reovirus particle (ISVP) that is associated with primary infection in the gut. Although LPS and PG alter reovirus thermostability, these bacterial envelope components did not affect reovirus utilization of its proteinaceous cellular receptor junctional adhesion molecule-A or cell entry kinetics. LPS and PG also did not affect the overall number of reovirus capsid proteins σ1 and σ3, suggesting their effect on virion thermostability is not mediated through altering the overall number of major capsid proteins on the virus. Incubation of reovirus with LPS and PG did not significantly affect the neutralizing efficiency of reovirus-specific antibodies. These data suggest that bacteria enhance reovirus infection of the intestinal tract by enhancing the thermal stability of the reovirus particle at a variety of temperatures through interactions between the viral particle and bacterial envelope components.


Asunto(s)
Bacillus subtilis/fisiología , Enterocitos/virología , Escherichia coli K12/fisiología , Infecciones por Reoviridae/virología , Reoviridae/fisiología , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Acetilglucosamina/toxicidad , Bacillus subtilis/metabolismo , Bacillus subtilis/ultraestructura , Bacillus subtilis/virología , Células CACO-2 , Endotoxinas/metabolismo , Endotoxinas/toxicidad , Enterocitos/efectos de los fármacos , Enterocitos/microbiología , Enterocitos/patología , Escherichia coli K12/metabolismo , Escherichia coli K12/ultraestructura , Escherichia coli K12/virología , Microbioma Gastrointestinal , Células HeLa , Calor , Humanos , Lipopolisacáridos/metabolismo , Lipopolisacáridos/toxicidad , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Electrónica de Transmisión , Peptidoglicano/metabolismo , Peptidoglicano/toxicidad , ARN/metabolismo , Estabilidad del ARN/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Reoviridae/química , Reoviridae/efectos de los fármacos , Reoviridae/patogenicidad , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/microbiología , Infecciones por Reoviridae/patología , Ácidos Teicoicos/metabolismo , Ácidos Teicoicos/toxicidad , Virión/química , Virión/patogenicidad , Virión/fisiología , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Proteína Fluorescente Roja
8.
Transgenic Res ; 28(1): 21-32, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30315482

RESUMEN

The alphacoronaviruses, transmissible gastroenteritis virus (TGEV) and Porcine epidemic diarrhea virus (PEDV) are sources of high morbidity and mortality in neonatal pigs, a consequence of dehydration caused by the infection and necrosis of enterocytes. The biological relevance of amino peptidase N (ANPEP) as a putative receptor for TGEV and PEDV in pigs was evaluated by using CRISPR/Cas9 to edit exon 2 of ANPEP resulting in a premature stop codon. Knockout pigs possessing the null ANPEP phenotype and age matched wild type pigs were challenged with either PEDV or TGEV. Fecal swabs were collected daily from each animal beginning 1 day prior to challenge with PEDV until the termination of the study. The presence of virus nucleic acid was determined by PCR. ANPEP null pigs did not support infection with TGEV, but retained susceptibility to infection with PEDV. Immunohistochemistry confirmed the presence of PEDV reactivity and absence of TGEV reactivity in the enterocytes lining the ileum in ANPEP null pigs. The different receptor requirements for TGEV and PEDV have important implications in the development of new genetic tools for the control of enteric disease in pigs.


Asunto(s)
Aminopeptidasas/genética , Animales Modificados Genéticamente/genética , Infecciones por Coronavirus/genética , Coronavirus/patogenicidad , Aminopeptidasas/deficiencia , Animales , Animales Modificados Genéticamente/virología , Sistemas CRISPR-Cas , Coronavirus/genética , Infecciones por Coronavirus/virología , Enterocitos/enzimología , Enterocitos/virología , Virus de la Diarrea Epidémica Porcina/patogenicidad , Porcinos , Virus de la Gastroenteritis Transmisible/patogenicidad
9.
Vet Res ; 50(1): 110, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856906

RESUMEN

Intestinal epithelium functions as a barrier to protect multicellular organisms from the outside world. It consists of epithelial cells closely connected by intercellular junctions, selective gates which control paracellular diffusion of solutes, ions and macromolecules across the epithelium and keep out pathogens. Rotavirus is one of the major enteric viruses causing severe diarrhea in humans and animals. It specifically infects the enterocytes on villi of small intestines. The polarity of rotavirus replication in their target enterocytes and the role of intestinal epithelial integrity were examined in the present study. Treatment with EGTA, a drug that chelates calcium and disrupts the intercellular junctions, (i) significantly enhanced the infection of rotavirus in primary enterocytes, (ii) increased the binding of rotavirus to enterocytes, but (iii) considerably blocked internalization of rotavirus. After internalization, rotavirus was resistant to EGTA treatment. To investigate the polarity of rotavirus infection, the primary enterocytes were cultured in a transwell system and infected with rotavirus at either the apical or the basolateral surface. Rotavirus preferentially infected enterocytes at the basolateral surface. Restriction of infection through apical inoculation was overcome by EGTA treatment. Overall, our findings demonstrate that integrity of the intestinal epithelium is crucial in the host's innate defense against rotavirus infection. In addition, the intercellular receptor is located basolaterally and disruption of intercellular junctions facilitates the binding of rotavirus to their receptor at the basolateral surface.


Asunto(s)
Enterocitos/virología , Células Epiteliales/virología , Mucosa Intestinal/citología , Rotavirus/clasificación , Rotavirus/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo/veterinaria , Ácido Egtácico/farmacología , Enterocitos/efectos de los fármacos , Miofibroblastos/fisiología , Porcinos , Internalización del Virus , Replicación Viral
10.
J Biol Chem ; 292(24): 10262-10274, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28455446

RESUMEN

Enterovirus 71 (EV71) has emerged as one of the most important enteroviruses since the eradication of poliovirus, and it causes severe neurological symptoms for which no effective antiviral drugs are available. Type I interferons (IFN) α/ß have been used clinically as antiviral therapy as the first line of defense against virus infections successfully for decades. However, treatment with type I interferons has not been effective in patients with EV71 infection. In this study, we found that in cells pretreated with IFN-ß, EV71 infection could still lead to a cytopathic effect, and the viral replication was not affected. The mechanism by which EV71 antagonizes interferon signaling, however, has been controversial. Our study indicated that EV71 infection did not inhibit phosphorylation of STAT1/2 induced by IFN-ß stimulation, but p-STAT1/2 transport into the nucleus was significantly blocked. We showed that EV71 infection reduced the formation of STAT/karyopherin-α1 (KPNA1) complex upon interferon stimulation and that the virus down-regulated the expression of KPNA1, a nuclear localization signal receptor for p-STAT1. Using specific caspase inhibitors and siRNA for caspase-3, we demonstrated that EV71 infection induced degradation of cellular KPNA1 in a caspase-3-dependent manner, which led to decreased induction of interferon-inducible genes and IFN response. Viral 2A and 3C proteases did not degrade KPNA1, inhibit the activity of ISRE or suppress the transcription of interferon-inducible genes induced by IFN-ß. Our study demonstrates a novel mechanism by which antiviral signaling is suppressed through degradation of KPNA1 by activated caspase-3 induced in an enteroviral infection.


Asunto(s)
Caspasa 3/metabolismo , Enterocitos/virología , Enterovirus Humano A/fisiología , Interferón beta/metabolismo , Janus Quinasa 1/metabolismo , Transducción de Señal , alfa Carioferinas/antagonistas & inhibidores , Transporte Activo de Núcleo Celular , Animales , Caspasa 3/química , Caspasa 3/genética , Chlorocebus aethiops , Enterocitos/inmunología , Enterocitos/metabolismo , Enterovirus Humano A/crecimiento & desarrollo , Células HT29 , Células HeLa , Humanos , Interferón beta/genética , Janus Quinasa 1/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Proteolisis , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Células Vero , Replicación Viral , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
11.
Intervirology ; 61(2): 53-63, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30176660

RESUMEN

Transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV) are similar coronaviruses, causing diseases characterized by vomiting, diarrhea, and death from severe dehydration in piglets. Thus, they have caused huge losses to the swine-breeding industry worldwide. Nowadays, they are easily transmitted among the continents via vehicles, equipment, and cargo. Both viruses establish an infection in porcine enterocytes in the small intestine, and their spike (S) proteins play a key role in the virus-cell binding process under unfavorable conditions when the intestine with a low pH is filled with a thick layer of mucus and proteases. Sialic acid, proteases, and low pH are three main inducers of coronavirus infection. However, the details of how sialic acid and low pH affect virus binding to the host cell are not determined, and the functions of the proteases are unknown. This review emphasizes the role of three factors in the invasion of TGEV and PEDV into porcine enterocytes and offers more insights into Alphacoronavirus infection in the intestinal environment.


Asunto(s)
Alphacoronavirus/fisiología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Enterocitos/virología , Interacciones Huésped-Patógeno , Alphacoronavirus/clasificación , Animales , Concentración de Iones de Hidrógeno , Ácido N-Acetilneuramínico/metabolismo , Péptido Hidrolasas/metabolismo , Filogenia , Virus de la Diarrea Epidémica Porcina/fisiología , Unión Proteica , Receptores Virales/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Relación Estructura-Actividad , Porcinos , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/virología , Virus de la Gastroenteritis Transmisible/fisiología
12.
Gastroenterol Hepatol ; 40(5): 339-347, 2017 May.
Artículo en Inglés, Español | MEDLINE | ID: mdl-28249699

RESUMEN

BACKGROUND: The life cycle of the hepatitis C virus (HCV) is closely associated with lipid metabolism. Recently, NPC1L1 (a cholesterol transporter) has been reported to function as an HCV receptor. This receptor is expressed in the hepatocyte canalicular membrane and in the intestine; serving as a key transporter for the cholesterol enterohepatic cycle. OBJECTIVES: We hypothesized that HCV might have a similar cycle, so we aimed to study the presence of HCV in bile and stools of infected patients. MATERIALS AND METHODS: Blood, feces, and duodenal bile samples were collected from patients infected with HCV. The biliary viral load was normalized to the bile salt concentration of each sample and the presence of HCV core protein was also evaluated. A total of 12 patients were recruited. HCV RNA was detected in the bile from ten patients. RESULTS: The mean viral load was 2.5log10IU/60mg bile salt. In the stool samples, HCV RNA was detected in ten patients (mean concentration 2.7log10IU/g of feces). CONCLUSIONS: HCV RNA is readily detectable and is present at relatively high concentrations in the bile and stool samples of infected patients. This may be relevant as a source of infection in men who have sex with men. Biliary HCV secretion may perhaps play a role in the persistence of viral infection via an enterohepatic cycle of the virus or intrahepatic spread.


Asunto(s)
Bilis/virología , Heces/virología , Hepacivirus/aislamiento & purificación , Hepatitis C Crónica/virología , Esparcimiento de Virus , Chile , Colesterol/sangre , Duodeno , Enterocitos/metabolismo , Enterocitos/virología , Circulación Enterohepática , Hepacivirus/crecimiento & desarrollo , Hepatitis C Crónica/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Mucosa Intestinal/metabolismo , Intestinos/virología , Estadios del Ciclo de Vida , Metabolismo de los Lípidos , Proteínas de la Membrana/fisiología , Proteínas de Transporte de Membrana , ARN Viral/análisis , Receptores Virales/fisiología , Triglicéridos/sangre , Carga Viral
14.
J Med Virol ; 88(11): 1837-43, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27110852

RESUMEN

Noroviruses constitute a family of ubiquitous and highly efficient human pathogens. In spite of decades of dedicated research, human noroviruses remain a major cause of gastroenteritis and severe diarrheal disease around the world. Recent findings have begun to unravel the complex mechanisms that regulate norovirus pathogenesis and persistent infection, including the important interplay between the virus, the host immune system, and commensal bacteria. Herein, we will summarize recent research developments regarding norovirus cell tropism, the use of M cells, and commensal bacteria to facilitate norovirus infection, and virus, host, and bacterial determinants of persistent norovirus infections. J. Med. Virol. 88:1837-1843, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Infecciones por Caliciviridae/virología , Enterocitos/virología , Gastroenteritis/virología , Norovirus/patogenicidad , Animales , Antivirales/uso terapéutico , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/microbiología , Técnicas de Cultivo de Célula , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Intestinos/virología , Ratones , Norovirus/fisiología , Simbiosis , Tropismo Viral , Replicación Viral
15.
Vet Pathol ; 53(1): 44-52, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26113613

RESUMEN

Porcine epidemic diarrhea virus (PEDV) was first recognized in North America in April 2013 and has since caused devastating disease. The objective of this study was to characterize disease and viral detection associated with an original North American PEDV isolate inoculated in neonatal piglets. Thirty-six 1-day-old cesarean-derived and colostrum-deprived piglets were randomly assigned to the control (n = 16) or challenged group (n = 20); the latter were orogastrically inoculated with 1 ml of US/Iowa/18984/2013 PEDV isolate titered at 1 × 10(3) plaque-forming units per milliliter. Rectal swabs were collected from all piglets prior to inoculation and every 12 hours postinoculation (hpi) thereafter, with 4 control and 5 challenged piglets euthanized at 12, 24, 48, and 72 hpi. One piglet had a positive real-time quantitative polymerase chain reaction test on rectal swab at 12 hpi, and all remaining piglets were positive thereafter, with highest viral quantities detected at 24 and 36 hpi. Diarrhea was evident in 30% and 100% of challenged piglets at 18 and 24 hpi, respectively. Viral antigen was detected in enterocytes by immunohistochemistry in the duodenum and ileum of piglets euthanized at 12 hpi and was apparent throughout the small intestine of all piglets thereafter, with villus height:crypt depth ratios consistently below 4:1. Viremia was confirmed in 18 of 20 pigs at euthanasia. Clinical disease was severe and developed rapidly following infection with an original North American PEDV isolate, with lesions, viremia, and antigen detection possible by 12 hpi.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Diarrea/veterinaria , Virus de la Diarrea Epidémica Porcina/aislamiento & purificación , Enfermedades de los Porcinos/patología , Animales , Antígenos Virales/análisis , Calostro/metabolismo , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Enterocitos/virología , Femenino , Inmunohistoquímica/veterinaria , Intestino Delgado/virología , Virus de la Diarrea Epidémica Porcina/patogenicidad , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Porcinos , Enfermedades de los Porcinos/virología
16.
Blood ; 120(3): 572-80, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22589473

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. In addition to blood transfusion and sexual transmission, HTLV-1 is transmitted mainly through prolonged breastfeeding, and such infection represents a major risk for the development of adult T-cell leukemia/lymphoma. Although HTLV-1-infected lymphocytes can be retrieved from maternal milk, the mechanisms of HTLV-1 transmission through the digestive tract remain unknown. In the present study, we assessed HTLV-1 transport across the epithelial barrier using an in vitro model. Our results show that the integrity of the epithelial barrier was maintained during coculture with HTLV-1-infected lymphocytes, because neither morphological nor functional alterations of the cell monolayer were observed. Enterocytes were not susceptible to HTLV-1 infection, but free infectious HTLV-1 virions could cross the epithelial barrier via a transcytosis mechanism. Such virions were able to infect productively human dendritic cells located beneath the epithelial barrier. Our data indicate that HTLV-1 crosses the tight epithelial barrier without disruption or infection of the epithelium to further infect target cells such as dendritic cells. The present study provides the first data pertaining to the mode of HTLV-1 transport across a tight epithelial barrier, as can occur during mother-to-child HTLV-1 transmission during breastfeeding.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/virología , Infecciones por HTLV-I/metabolismo , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Transcitosis/fisiología , Virión/metabolismo , Células CACO-2 , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Enterocitos/virología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células HEK293 , Células HT29 , Infecciones por HTLV-I/transmisión , Infecciones por HTLV-I/virología , Humanos , Microscopía Electrónica de Transmisión , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/virología , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Uniones Estrechas/virología
17.
Proteomics ; 13(7): 1121-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23303713

RESUMEN

Rotavirus (RV) infection is the main cause of acute dehydrating diarrhea in infants and young children below 5 years old worldwide. RV infection causes a global shutoff of host proteins as many other viruses do. However, previous studies revealed that RV could selectively upregulated the expression of some host proteins that then played important roles in RV infection. To globally explor such host proteins that were upregulated in early human rotavirus (HRV) infection, proteomic methods were used and a total of ten upregulated host proteins were unambiguously identified. Cyclophilin A (CYPA), a peptidyl-prolyl cis-trans isomerase, was among these upregulated host proteins. Following infection, CYPA was recruited to the viroplasm and interacted with HRV structural protein VP2; CYPA reduced host susceptibility to HRV infection and inhibited replication of HRV by repressing the expression of viral proteins. Furthermore, we found that the increased expression of CYPA in enterocytes of small intestine correlated to the period when BALB/c mice became resistant to RV diarrhea. Together, we identified CYPA as a novel host restriction factor that confered protection against RV infection and might contribute to host susceptibility to RV diarrhea.


Asunto(s)
Ciclofilina A/metabolismo , Interacciones Huésped-Patógeno , Proteómica/métodos , Infecciones por Rotavirus/metabolismo , Infecciones por Rotavirus/virología , Rotavirus/fisiología , Animales , Células CACO-2 , Proteínas de la Cápside/metabolismo , Diarrea/metabolismo , Diarrea/patología , Diarrea/virología , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados , Infecciones por Rotavirus/inmunología , Regulación hacia Arriba , Replicación Viral
18.
Gastroenterology ; 141(4): 1220-30, 1230.e1-3, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21726511

RESUMEN

BACKGROUND & AIMS: Chronic infection with hepatitis B or C virus (HBV or HCV) is a leading cause of cirrhosis by unknown mechanisms of pathogenesis. Translocation of gut microbial products into the systemic circulation might increase because of increased intestinal permeability, bacterial overgrowth, or impaired clearance of microbial products by Kupffer cells. We investigated whether the extent and progression of liver disease in patients with chronic HBV or HCV infection are associated with microbial translocation and subsequent activation of monocytes. METHODS: In a retrospective study, we analyzed data from 16 patients with minimal fibrosis, 68 with cirrhosis, and 67 uninfected volunteers. We analyzed plasma levels of soluble CD14 (sCD14), intestinal fatty acid binding protein, and interleukin-6 by enzyme-linked immunosorbent assay, and lipopolysaccharide (LPS) by the limulus amebocyte lysate assay, at presentation and after antiviral treatment. RESULTS: Compared with uninfected individuals, HCV- and HBV-infected individuals had higher plasma levels of LPS, intestinal fatty acid binding protein (indicating enterocyte death), sCD14 (produced upon LPS activation of monocytes), and interleukin-6. Portal hypertension, indicated by low platelet counts, was associated with enterocyte death (P=.045 at presentation, P<.0001 after therapy). Levels of sCD14 correlated with markers of hepatic inflammation (P=.02 for aspartate aminotransferase, P=.002 for ferritin) and fibrosis (P<.0001 for γ-glutamyl transpeptidase, P=.01 for alkaline phosphatase, P<.0001 for α-fetoprotein). Compared to subjects with minimal fibrosis, subjects with severe fibrosis at presentation had higher plasma levels of sCD14 (P=.01) and more hepatic CD14+ cells (P=.0002); each increased risk for disease progression (P=.0009 and P=.005, respectively). CONCLUSIONS: LPS-induced local and systemic inflammation is associated with cirrhosis and predicts progression to end-stage liver disease in patients with HBV or HCV infection.


Asunto(s)
Traslocación Bacteriana , Hepatitis B Crónica/complicaciones , Hepatitis C Crónica/complicaciones , Interacciones Huésped-Patógeno , Intestinos/virología , Cirrosis Hepática/virología , Monocitos/virología , Biomarcadores/sangre , Biopsia , Muerte Celular , Progresión de la Enfermedad , Enfermedad Hepática en Estado Terminal/microbiología , Enfermedad Hepática en Estado Terminal/virología , Enterocitos/microbiología , Enterocitos/patología , Enterocitos/virología , Ensayo de Inmunoadsorción Enzimática , Proteínas de Unión a Ácidos Grasos/sangre , Femenino , Hepatitis B Crónica/diagnóstico , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/microbiología , Hepatitis C Crónica/diagnóstico , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/microbiología , Humanos , Hipertensión Portal/microbiología , Hipertensión Portal/virología , Interleucina-6/sangre , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Macrófagos del Hígado/microbiología , Macrófagos del Hígado/virología , Prueba de Limulus , Receptores de Lipopolisacáridos/sangre , Lipopolisacáridos/sangre , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/inmunología , Cirrosis Hepática/microbiología , Modelos Logísticos , Masculino , Maryland , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/microbiología , Oportunidad Relativa , Estudios Retrospectivos , Índice de Severidad de la Enfermedad
19.
J Prev Med Hyg ; 53(3): 165-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23362623

RESUMEN

INTRODUCTION: The interest grown in these years about emerging pathogens in the onset of intestinal disease showed that the pathogenic mechanism is a multifactorial event. Our objective was to evaluate the role of co-infection with rotavirus in the expression of Aeromonas spp adhesiveness. METHODS: The rate of co-infection involves contact of Caco-2 cells with the virus, followed by adsorption for 1 and 2 hours. Aliquots of bacterial suspensions were added to tissue-culture plates. After infection, cell monolayers were lysed; serially diluted lysates were plated to determine the number of bound bacteria by performing colony forming units (CFU) counts. RESULTS: Non-adhesive strains were not subject to variations resulting from co-infection, while those who had medium or high adhesiveness gave rise to an increase of the same. DISCUSSION: Infection with rotavirus promotes the Aeromonas ability to adhere to Caco-2 cells and this effect depends on the duration of infection and on the starting adhesiveness of bacteria strain.


Asunto(s)
Aeromonas hydrophila/fisiología , Adhesión Bacteriana/fisiología , Células CACO-2/microbiología , Coinfección/microbiología , Enterocitos/microbiología , Enterocitos/virología , Rotavirus/patogenicidad , Humanos , Rotavirus/fisiología
20.
Nat Commun ; 13(1): 17, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013162

RESUMEN

Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Caenorhabditis elegans , Enterocitos , Interacciones Huésped-Patógeno/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Caenorhabditis elegans/inmunología , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/virología , Proteínas de Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/metabolismo , Enterocitos/inmunología , Enterocitos/microbiología , Enterocitos/virología , Inmunidad Innata/fisiología , Intestinos/microbiología , Intestinos/virología , Invertebrados/inmunología , Microsporidios/patogenicidad , Virus ARN/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA