Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 181(7): 1475-1488.e12, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32479746

RESUMEN

Viruses are a constant threat to global health as highlighted by the current COVID-19 pandemic. Currently, lack of data underlying how the human host interacts with viruses, including the SARS-CoV-2 virus, limits effective therapeutic intervention. We introduce Viral-Track, a computational method that globally scans unmapped single-cell RNA sequencing (scRNA-seq) data for the presence of viral RNA, enabling transcriptional cell sorting of infected versus bystander cells. We demonstrate the sensitivity and specificity of Viral-Track to systematically detect viruses from multiple models of infection, including hepatitis B virus, in an unsupervised manner. Applying Viral-Track to bronchoalveloar-lavage samples from severe and mild COVID-19 patients reveals a dramatic impact of the virus on the immune system of severe patients compared to mild cases. Viral-Track detects an unexpected co-infection of the human metapneumovirus, present mainly in monocytes perturbed in type-I interferon (IFN)-signaling. Viral-Track provides a robust technology for dissecting the mechanisms of viral-infection and pathology.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Interacciones Huésped-Patógeno , Neumonía Viral/fisiopatología , Programas Informáticos , Animales , Betacoronavirus/aislamiento & purificación , COVID-19 , Coinfección/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Humanos , Interferones/inmunología , Pulmón/patología , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2 , Sensibilidad y Especificidad , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual
2.
Nature ; 584(7819): 115-119, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32454513

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents a global health emergency that is in urgent need of intervention1-3. The entry of SARS-CoV-2 into its target cells depends on binding between the receptor-binding domain (RBD) of the viral spike protein and its cellular receptor, angiotensin-converting enzyme 2 (ACE2)2,4-6. Here we report the isolation and characterization of 206 RBD-specific monoclonal antibodies derived from single B cells from 8 individuals infected with SARS-CoV-2. We identified antibodies that potently neutralize SARS-CoV-2; this activity correlates with competition with ACE2 for binding to RBD. Unexpectedly, the anti-SARS-CoV-2 antibodies and the infected plasma did not cross-react with the RBDs of SARS-CoV or Middle East respiratory syndrome-related coronavirus (MERS-CoV), although there was substantial plasma cross-reactivity to their trimeric spike proteins. Analysis of the crystal structure of RBD-bound antibody revealed that steric hindrance inhibits viral engagement with ACE2, thereby blocking viral entry. These findings suggest that anti-RBD antibodies are largely viral-species-specific inhibitors. The antibodies identified here may be candidates for development of clinical interventions against SARS-CoV-2.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Neumonía Viral/inmunología , Neumonía Viral/virología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto , Anciano , Enzima Convertidora de Angiotensina 2 , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Linfocitos B/citología , Linfocitos B/inmunología , Betacoronavirus/química , COVID-19 , Niño , Células Clonales/citología , Células Clonales/inmunología , Reacciones Cruzadas , Cristalización , Cristalografía por Rayos X , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Pruebas de Neutralización , Pandemias , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Plasma/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo
3.
Antimicrob Agents Chemother ; : e0005424, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687016

RESUMEN

Human enteroviruses are the major pathogens causing hand-foot-and-mouth disease in infants and young children throughout the world, and infection with enterovirus is also associated with severe complications, such as aseptic meningitis and myocarditis. However, there are no antiviral drugs available to treat enteroviruses infection at present. In this study, we found that 4'-fluorouridine (4'-FlU), a nucleoside analog with low cytotoxicity, exhibited broad-spectrum activity against infections of multiple enteroviruses with EC50 values at low micromolar levels, including coxsackievirus A10 (CV-A10), CV-A16, CV-A6, CV-A7, CV-B3, enterovirus A71 (EV-A71), EV-A89, EV-D68, and echovirus 6. With further investigation, the results indicated that 4'-FlU directly interacted with the RNA-dependent RNA polymerase of enterovirus, the 3D pol, and impaired the polymerase activity of 3D pol, hence inhibiting viral RNA synthesis and significantly suppressing viral replication. Our findings suggest that 4'-FlU could be promisingly developed as a broad-spectrum direct-acting antiviral agent for anti-enteroviruses therapy.

4.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34873039

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), binds to host receptor angiotensin-converting enzyme 2 (ACE2) through its spike (S) glycoprotein, which mediates membrane fusion and viral entry. However, the expression of ACE2 is extremely low in a variety of human tissues, especially in the airways. Thus, other coreceptors and/or cofactors on the surface of host cells may contribute to SARS-CoV-2 infection. Here, we identified nonmuscle myosin heavy chain IIA (MYH9) as an important host factor for SARS-CoV-2 infection of human pulmonary cells by using APEX2 proximity-labeling techniques. Genetic ablation of MYH9 significantly reduced SARS-CoV-2 pseudovirus infection in wild type (WT) A549 and Calu-3 cells, and overexpression of MYH9 enhanced the pseudovirus infection in WT A549 and H1299 cells. MYH9 was colocalized with the SARS-CoV-2 S and directly interacted with SARS-CoV-2 S through the S2 subunit and S1-NTD (N-terminal domain) by its C-terminal domain (designated as PRA). Further experiments suggested that endosomal or myosin inhibitors effectively block the viral entry of SARS-CoV-2 into PRA-A549 cells, while transmembrane protease serine 2 (TMPRSS2) and cathepsin B and L (CatB/L) inhibitors do not, indicating that MYH9 promotes SARS-CoV-2 endocytosis and bypasses TMPRSS2 and CatB/L pathway. Finally, we demonstrated that loss of MYH9 reduces authentic SARS-CoV-2 infection in Calu-3, ACE2-A549, and ACE2-H1299 cells. Together, our results suggest that MYH9 is a candidate host factor for SARS-CoV-2, which mediates the virus entering host cells by endocytosis in an ACE2-dependent manner, and may serve as a potential target for future clinical intervention strategies.


Asunto(s)
COVID-19/virología , Cadenas Pesadas de Miosina/metabolismo , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Línea Celular , Membrana Celular/metabolismo , Humanos , Pulmón/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Unión Proteica , Dominios Proteicos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
5.
Proteomics ; 23(2): e2200362, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36254857

RESUMEN

Enterovirus A71 (EV71) infection can cause hand, foot, and mouth disease (HFMD) and severe neurological complications in children. However, the biological processes regulated by EV71 remain poorly understood. Herein, proteomics and metabonomics studies were conducted to uncover the mechanism of EV71 infection in rhabdomyosarcoma (RD) cells and identify potential drug targets. Differential expressed proteins from enriched membrane were analyzed by isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics technology. Twenty-six differential proteins with 1.5-fold (p < 0.05) change were detected, including 14 upregulated proteins and 12 downregulated proteins. The upregulated proteins are mainly involved in metabolic process, especially in the glycolysis pathway. Alpha-enolase (ENO1) protein was found to increase with temporal dependence following EV71 infection. The targeted metabolomics analysis revealed that glucose absorption and glycolysis metabolites were increased after EV71 infection. The glycolysis pathway was inhibited by knocking down ENO1 or the use of a glycolysis inhibitor (dichloroacetic acid [DCA]); and we found that EV71 infection was inhibited by depleting ENO1 or using DCA. Our study indicates that EV71 may reprogram glucose metabolism by activating glycolysis, and EV71 infection can be inhibited by interrupting the glycolysis pathway. ENO1 may be a potential target against EV71, and DCA could act as an inhibitor of EV71.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Niño , Humanos , Enterovirus/metabolismo , Enterovirus Humano A/metabolismo , Proteómica , Infecciones por Enterovirus/metabolismo , Proteínas/metabolismo , Metabolómica , Redes y Vías Metabólicas
6.
Am J Transplant ; 23(7): 946-956, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37084847

RESUMEN

Induced regulatory T (iTreg) cells play a vital role in immune tolerance and in controlling chronic inflammation. Generated in the periphery, iTreg cells are suitable for responding to alloantigens and preventing transplant rejection. Nevertheless, their clinical application has been impeded by the plasticity and instability attributed to the loss of forkhead box protein 3 expression, raising concerns that iTreg may be converted to effector T cells and even exert a pathogenic effect. Herein, second-generation short hairpin RNAs loaded with 3 pairs of small interfering RNAs were utilized to target the T-box transcription factor TBX21. In addition, 2 immunosuppressive cytokines, namely, transforming growth factor beta and interleukin 10, were constitutively expressed. This novel engineering strategy allowed the generation of stably induced regulatory T (SI Treg) cells, which maintained the expression of forkhead box protein 3 even in an unfavorable environment and exerted potent immunosuppressive functions in vitro. Furthermore, SI Treg cells demonstrated an effector transcriptional profile. Finally, SI Treg cells showed a significant protective effect against graft-versus-host disease-related deaths in a xenotransplantation model. Collectively, these results signify that SI Treg cells hold great promise for future clinical application and offer a rational therapeutic approach for transplant rejection.


Asunto(s)
Enfermedad Injerto contra Huésped , Linfocitos T Reguladores , Humanos , Citocinas/metabolismo , Expresión Génica Ectópica , Enfermedad Injerto contra Huésped/prevención & control , Factores de Transcripción Forkhead/metabolismo
7.
J Immunol ; 206(12): 2900-2908, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34049969

RESUMEN

The relatively low partial pressure of oxygen, reduced oxygen saturation, and aberrant plasma metabolites in COVID-19 may alter energy metabolism in peripheral immune cells. However, little is known regarding the immunometabolic defects of T cells in COVID-19 patients, which may contribute to the deregulated immune functions of these cells. In this study, we longitudinally characterized the metabolic profiles of resting and activated T cells from acutely infected and convalescent COVID-19 patients by flow cytometry and confirmed the metabolic profiles with a Seahorse analyzer. Non-COVID-19 and healthy subjects were enrolled as controls. We found that ex vivo T cells from acutely infected COVID-19 patients were highly activated and apoptotic and displayed more extensive mitochondrial metabolic dysfunction, especially cells in CD8+ T cell lineages, than those from convalescent COVID-19 patients or healthy controls, but slightly disturbed mitochondrial metabolic activity was observed in non-COVID-19 patients. Importantly, plasma IL-6 and C-reactive protein (CRP) levels positively correlated with mitochondrial mass and negatively correlated with fatty acid uptake in T cells from COVID-19 patients. Additionally, compared with those from healthy controls, in vitro-activated T cells from acutely infected COVID-19 patients showed signs of lower glycolysis, a reduced glycolytic capacity, and a decreased glycolytic reserve, accompanied by lower activation of mTOR signaling. Thus, newly identified defects in T cell mitochondrial metabolic functions and metabolic reprogramming upon activation might contribute to immune deficiency in COVID-19.


Asunto(s)
COVID-19 , Linfocitos T CD8-positivos , Glucólisis , Humanos , Saturación de Oxígeno , SARS-CoV-2
8.
J Immunol ; 207(7): 1848-1856, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34452933

RESUMEN

Immune cell responses are strikingly altered in patients with severe coronavirus disease 2019 (COVID-19), but the immunoregulatory process in these individuals is not fully understood. In this study, 23 patients with mild and 22 patients with severe COVID-19 and 6 asymptomatic carriers of COVID-19 were enrolled, along with 44 healthy controls (HC). Peripheral immune cells in HC and patients with COVID-19 were comprehensively profiled using mass cytometry. We found that in patients with severe COVID-19, the number of HLA-DRlow/- monocytes was significantly increased, but that of mucosal-associated invariant T (MAIT) cells was greatly reduced. MAIT cells were highly activated but functionally impaired in response to Escherichia coli and IL-12/IL-18 stimulation in patients with severe COVID-19, especially those with microbial coinfection. Single-cell transcriptome analysis revealed that IFN-stimulated genes were significantly upregulated in peripheral MAIT cells and monocytes from patients with severe COVID-19. IFN-α pretreatment suppressed MAIT cells' response to E. coli by triggering high levels of IL-10 production by HLA-DRlow/--suppressive monocytes. Blocking IFN-α or IL-10 receptors rescued MAIT cell function in patients with severe COVID-19. Moreover, plasma from patients with severe COVID-19 inhibited HLA-DR expression by monocytes through IL-10. These data indicate a unique pattern of immune dysregulation in severe COVID-19, which is characterized by enrichment of suppressive HLA-DRlow/- monocytes associated with functional impairment of MAIT cells through the IFN/IL-10 pathway.


Asunto(s)
COVID-19/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , Interleucina-10/metabolismo , Monocitos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , SARS-CoV-2/fisiología , Adolescente , Adulto , Enfermedades Asintomáticas , Células Cultivadas , Niño , Coinfección , Progresión de la Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Adulto Joven
9.
J Gen Virol ; 101(11): 1145-1155, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32762804

RESUMEN

The species Enterovirus A (EV-A) consists of two conventional clusters and one unconventional cluster. At present, sequence analysis shows no evidence of recombination between conventional and unconventional EV-A types. However, the factors underlying this genetic barrier are unclear. Here, we systematically dissected the genome components linked to these peculiar phenomena, using the viral reverse genetic tools. We reported that viral capsids of the unconventional EV-A types expressed poorly in human cells. The trans-encapsidation outputs across conventional and unconventional EV-A types were also with low efficiency. However, replicons of conventional types bearing exchanged 5'-untranslated region (UTR) or non-structural regions from the unconventional types were replication-competent. Furthermore, we created a viable recombinant EVA71 (conventional type) with its P3 region replaced by that from EVA89 (unconventional type). Thus, our data for the first time reveal the potential for fertile genetic exchanges between conventional and unconventional EV-A types. It also discloses that the mysterious recombination barriers may lie in uncoordinated capsid expression and particle assembly by different EV-A clusters.


Asunto(s)
Enterovirus Humano A/genética , Recombinación Genética , Regiones no Traducidas 5' , Animales , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Línea Celular Tumoral , Enterovirus Humano A/clasificación , Enterovirus Humano A/fisiología , Expresión Génica , Genoma Viral , Humanos , Filogenia , Replicón , Replicación Viral
10.
Biochem Biophys Res Commun ; 526(1): 135-140, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32199615

RESUMEN

The new coronavirus (SARS-CoV-2) outbreak from December 2019 in Wuhan, Hubei, China, has been declared a global public health emergency. Angiotensin I converting enzyme 2 (ACE2), is the host receptor by SARS-CoV-2 to infect human cells. Although ACE2 is reported to be expressed in lung, liver, stomach, ileum, kidney and colon, its expressing levels are rather low, especially in the lung. SARS-CoV-2 may use co-receptors/auxiliary proteins as ACE2 partner to facilitate the virus entry. To identify the potential candidates, we explored the single cell gene expression atlas including 119 cell types of 13 human tissues and analyzed the single cell co-expression spectrum of 51 reported RNA virus receptors and 400 other membrane proteins. Consistent with other recent reports, we confirmed that ACE2 was mainly expressed in lung AT2, liver cholangiocyte, colon colonocytes, esophagus keratinocytes, ileum ECs, rectum ECs, stomach epithelial cells, and kidney proximal tubules. Intriguingly, we found that the candidate co-receptors, manifesting the most similar expression patterns with ACE2 across 13 human tissues, are all peptidases, including ANPEP, DPP4 and ENPEP. Among them, ANPEP and DPP4 are the known receptors for human CoVs, suggesting ENPEP as another potential receptor for human CoVs. We also conducted "CellPhoneDB" analysis to understand the cell crosstalk between CoV-targets and their surrounding cells across different tissues. We found that macrophages frequently communicate with the CoVs targets through chemokine and phagocytosis signaling, highlighting the importance of tissue macrophages in immune defense and immune pathogenesis.


Asunto(s)
Betacoronavirus/fisiología , Receptores Virales/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Enzima Convertidora de Angiotensina 2 , COVID-19 , Coronavirus , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Humanos , Macrófagos/metabolismo , Especificidad de Órganos , Pandemias , Péptido Hidrolasas/genética , Péptido Hidrolasas/aislamiento & purificación , Peptidil-Dipeptidasa A/genética , Neumonía Viral/inmunología , Neumonía Viral/virología , Receptores Virales/aislamiento & purificación , SARS-CoV-2
11.
Biochem Biophys Res Commun ; 529(1): 97-103, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32560826

RESUMEN

Enterovirus A71 (EVA71) and Coxsackievirus A10 (CVA10) are representative types of Enterovirus A. Dependent on the host cell types, the EVA71 entry may utilize clathrin-, caveola-, and endophilin-A2-mediated endocytosis. However, the cell-entry and intracellular trafficking pathways of CVA10, using KREMEN1 as its receptor, are unclear. Here, we tested the relevant mechanisms through RNA interference (RNAi) and chemical inhibitors. We found that endocytosis of EVA71 and CVA10 in rhabdomyosarcoma (RD) cells engaged multiple pathways, and both viruses required Rac1. Interestingly, while CDC42 and Pak1 participated in EVA71 infection, PI3K played a role in CVA10 infection. The functions of Rab proteins in intracellular trafficking of CVA10 and EVA71 were examined by RNAi. Knockdown of Rab5 and Rab21 significantly reduced CVA10 infectivity, while knockdown of Rab5, Rab7 and Rab9 reduced EVA71 infectivity. Confocal microscopy confirmed the colocalization of CVA10 virions with Rab5 or Rab21, and colocalization of EVA71 virions with Rab5 or Rab7. Additionally, we observed that both CVA10 and EVA71 infections were inhibited by endosome acidification inhibitors, bafilomycin-A1 and NH4Cl. Together, our findings comparatively illustrate the entry and intracellular trafficking processes of representative Enterovirus A types and revealed novel enterovirus intervention targets.


Asunto(s)
Enterovirus Humano A/fisiología , Enterovirus Humano A/patogenicidad , Enterovirus/fisiología , Enterovirus/patogenicidad , Proteínas de Unión al GTP rab5/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Línea Celular Tumoral , Infecciones por Coxsackievirus/etiología , Infecciones por Coxsackievirus/virología , Endocitosis/fisiología , Endosomas/metabolismo , Infecciones por Enterovirus/etiología , Infecciones por Enterovirus/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Virulencia/fisiología , Internalización del Virus , Proteínas de Unión al GTP rab5/antagonistas & inhibidores , Proteínas de Unión al GTP rab5/genética
12.
Nanotechnology ; 31(11): 115101, 2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766038

RESUMEN

Disulfiram (DSF), an FDA approved drug for the treatment of alcoholism, has shown its effectiveness against diverse cancer types. Thus, we developed a disulfiram-loaded scaffold using the electrospinning method to enhance the stability of DSF and to facilitate its appropriate distribution to tumor tissues. The drug release profile of the disulfiram-loaded scaffold was examined by high-performance liquid chromatography. We obtained mechanical and morphological characterizations of A549 cells treated with different scaffolds by various techniques to evaluate its antitumor properties. This work revealed that the cells after the treatment with the disulfiram-loaded scaffold exhibited a lower height and a larger elastic modulus compared with the untreated cells and those treated with the neat electrospun fibers. The changes were the indicators of cell apoptosis. Taken collectively, the results indicate that DSF was successfully incorporated into the electrospun fibers, and the disulfiram-loaded scaffold has great potential for inhibiting the regional recurrence of cancer.


Asunto(s)
Disulfiram/química , Nanofibras/química , Polivinilos/química , Células A549 , Apoptosis/efectos de los fármacos , Disulfiram/metabolismo , Disulfiram/farmacología , Portadores de Fármacos/química , Liberación de Fármacos , Módulo de Elasticidad , Humanos , Microscopía de Fuerza Atómica
13.
Virol J ; 15(1): 1, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29298696

RESUMEN

BACKGROUND: Human enterovirus 71 (EV71) was previously known to enter cells through clathrin or caveolar mediated endocytic pathways. However, we observed chlorpromazine (CPZ) or dynasore (DNS), which inhibit clathrin and dynamin mediated endocytosis, did not suppress EV71 cell entry in particular cell types. So the current knowledge of entry mechanisms by EV71 is not complete. METHODS: Viral infection was examined by flow cytometry or end-point dilution assays. Viral entry was monitored by immunofluorescence or pseudoviral infections. Various inhibitors were utilized for manipulating endocytic pathways. Cellular proteins were knockdown by siRNA. RESULTS: CPZ and DNS did not inhibit but rather enhance viral infection in A549 cells, while they inhibited infections in other cells tested. We further found CPZ did not affect EV71 binding to target cells and failed to affect viral translation and replication, but enhanced viral entry in A549 cells. Immunofluorescence microscopy further confirmed this increased entry. Using siRNA experiment, we found that the enhancement of EV71 infection by CPZ did not require the components of clathrin mediated endocytosis. Finally, CPZ also enhanced infection by Coxackivirus A16 in A549 cells. CONCLUSIONS: CPZ and DNS, previously reported as EV71 entry inhibitors, may rather lead to increased viral infection in particular cell types. CPZ and DNS increased viral entry and not other steps of viral life cycles. Therefore, our study indicated an unknown dynamin-independent entry pathway utilized by enteroviruses that cause Hand-Foot-and-Mouth Diseases.


Asunto(s)
Endocitosis/efectos de los fármacos , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/virología , Enfermedad de Boca, Mano y Pie/virología , Internalización del Virus/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Clorpromazina/farmacología , Clatrina/metabolismo , Dinaminas/metabolismo , Infecciones por Enterovirus/metabolismo , Regulación Viral de la Expresión Génica/efectos de los fármacos , Enfermedad de Boca, Mano y Pie/metabolismo , Humanos , Hidrazonas/farmacología , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
Nanotechnology ; 29(43): 435701, 2018 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-30047924

RESUMEN

The silver nanowire (AgNW) transparent electrode is one of the promising components for flexible electronics due to its high electrical and thermal conductivity, optical transparency and flexibility. However, the application of the AgNW electrode with an improved performance is generally limited by its poor long-term stability. As the name suggests, the transparent electrode is usually exposed to visible light in various applications. Unlike other electrode materials, AgNWs show unique and complicated behavior under long-term visible light illumination. In this study, the comprehensive effect of visible light irradiation on the AgNW transparent electrode is initially investigated in detail. Light irradiation induces the migration of silver to enhance the nanowire contacts while also leading to the generation and growth of particles and diameter loss in the nanowire. Light irradiation accelerates the sulfidation and oxidation of the AgNWs as well, resulting in the emergence of degradation products on the nanowire surface. All these effects influence the sheet resistance of the AgNW electrode during light illumination. The light-induced change of sheet resistance also relates to the nanowire concentration due to the sensitivity of the wire-wire contact resistance near the percolation threshold. It is believed that this work will be a valuable reference for the design, processing and application of transparent electrodes used in numerous optoelectronic devices.

15.
Gastroenterology ; 144(2): 414-425.e7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23089201

RESUMEN

BACKGROUND & AIMS: The type III interferons (IFN-λs: interleukin [IL]-28a, IL-28b, and IL-29) have important roles in hepatitis C virus (HCV) infection, but little is understood about what cells produce these cytokines or how production is activated. We investigated whether human immune cells recognize HCV-infected cells and respond by producing IFN-λ. METHODS: We cultured healthy human peripheral blood mononuclear cells (PBMCs) with different populations of immune cells and Japanese fulminant hepatitis-1 (JFH-1) HCV-infected Huh7.5 (cell culture-derived HCV particles [HCVcc]/Huh7.5) cells. RESULTS: Human PBMCs recognized HCVcc/Huh7.5 cells and responded by producing IFN-α, IFN-γ, and IFN-λ. A rare subset of myeloid dendritic cells (mDCs), which are blood DC antigen (BDCA)+ (also called mDC2 cells), were the major source of IL-28 and IL-29 production in response to HCVcc/Huh7.5 cells. Plasmacytoid DCs produced IFN-α, whereas natural killer and natural killer T cells were the main source of IFN-γ production in co-culture experiments. Of the endosomal Toll-like receptors (TLRs)3, 7, 8, and 9, only TLR3 or double-stranded HCV RNA induced production of IL-28 and IL-29 by mDC2s; endosomal maturation was required. Production of IFN-α and IFN-λ were linked-IFN-λ increased production of IFN-α by plasmacytoid DCs and IFN-α significantly increased production of IFN-λ. CONCLUSIONS: mDC2s are a major source of IFN-λ production by PBMCs in response to HCVcc/Huh7.5 cells. mDC2s are activated through the TLR3 pathway, indicating that human DCs efficiently can initiate an immune response against HCV infection. IFN-λ therefore has an important role in HCV infection.


Asunto(s)
Células Dendríticas/metabolismo , Hepacivirus/inmunología , Anticuerpos contra la Hepatitis C/análisis , Hepatitis C Crónica/inmunología , Interferón-alfa/biosíntesis , Interferón gamma/biosíntesis , Células Mieloides/metabolismo , Antivirales , Células Cultivadas , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/virología , Humanos , Inmunidad Celular , Interferones , Interleucinas/biosíntesis , Células Mieloides/inmunología , ARN Viral/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Hepatology ; 58(3): 940-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22577054

RESUMEN

UNLABELLED: Recognition of hepatitis C virus (HCV)-infected hepatocyes and interferon (IFN) induction are critical in antiviral immune response. We hypothesized that cell-cell contact between plasmacytoid dendritic cells (pDCs) and HCV-infected cells was required for IFN-α induction through the involvement of cell-surface molecules. Coculture of human peripheral blood mononuclear cells (PBMCs) with genotype 1a full-length (FL) HCV genomic replicon cells or genotype 2a Japanese fulminant hepatitis type 1 (JFH-1) virus-infected hepatoma cells (JFH-1), and not with uninfected hepatoma cells (Huh7.5), induced IFN-α production. Depletion of pDCs from PBMCs attenuated IFN-α release, and purified pDCs produced high levels of IFN-α after coculture with FL replicons or JFH-1-infected cells. IFN-α induction by HCV-containing hepatoma cells required viral replication, direct cell-cell contact with pDCs, and receptor-mediated endocytosis. We determined that the tetraspanin proteins, CD81 and CD9, and not other HCV entry receptors, were required for IFN-α induction in pDCs by HCV-infected hepatoma cells. Disruption of cholesterol-rich membrane microdomains, the localization site of CD81, or inhibition of the CD81 downstream molecule, Rac GTPase, inhibited IFN-α production. IFN-α induction involved HCV RNA and Toll-like receptor (TLR) 7. IFN-α production by HCV-infected hepatoma cells was decreased in pDCs from HCV-infected patients, compared to healthy controls. We found that preexposure of healthy PBMCs to HCV viral particles attenuated IFN-α induction by HCV-infected hepatoma cells or TLR ligands, and this inhibitory effect could be prevented by an anti-HCV envelope glycoprotein 2-blocking antibody. CONCLUSION: Our novel data show that recognition of HCV-infected hepatoma cells by pDCs involves CD81- and CD9-associated membrane microdomains and induces potent IFN-α production.


Asunto(s)
Células Dendríticas/patología , Hepacivirus/aislamiento & purificación , Hepatocitos/metabolismo , Hepatocitos/patología , Interferón-alfa/metabolismo , Tetraspanina 28/fisiología , Tetraspanina 29/fisiología , Tetraspaninas/fisiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Estudios de Casos y Controles , Comunicación Celular/fisiología , Línea Celular Tumoral , Técnicas de Cocultivo , Células Dendríticas/fisiología , Endocitosis/fisiología , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatitis C/virología , Hepatocitos/virología , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Microdominios de Membrana/fisiología , Replicación Viral/fisiología
17.
Microbiol Spectr ; 12(4): e0333223, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38441464

RESUMEN

Enterovirus A89 (EV-A89) is an unconventional strain belonging to the Enterovirus A species. Limited research has been conducted on EV-A89, leaving its biological and pathogenic properties unclear. Developing reverse genetic tools for EV-A89 would help to unravel its infection mechanisms and aid in the development of vaccines and anti-viral drugs. In this study, an infectious clone for EV-A89 was successfully constructed and recombinant enterovirus A89 (rEV-A89) was generated. The rEV-A89 exhibited similar characteristics such as growth curve, plaque morphology, and dsRNA expression with parental strain. Four amino acid substitutions were identified in the EV-A89 capsid, which were found to enhance viral infection. Mechanistic studies revealed that these substitutions increased the virus's cell-binding ability. Establishing reverse genetic tools for EV-A89 will significantly contribute to understanding viral infection and developing anti-viral strategies.IMPORTANCEEnterovirus A species contain many human pathogens and have been classified into conventional cluster and unconventional cluster. Most of the research focuses on various conventional members, while understanding of the life cycle and infection characteristics of unconventional viruses is still very limited. In our study, we constructed the infectious cDNA clone and single-round infectious particles for the unconventional EV-A89, allowing us to investigate the biological properties of recombinant viruses. Moreover, we identified key amino acids residues that facilitate EV-A89 infection and elucidate their roles in enhancing viral binding to host cells. The establishment of the reverse genetics system will greatly facilitate future study on the life cycle of EV-A89 and contribute to the development of prophylactic vaccines and anti-viral drugs.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Vacunas , Humanos , Enterovirus/genética , Enterovirus Humano A/genética , Antígenos Virales , Sustitución de Aminoácidos , Células Clonales , Antivirales/farmacología
18.
Virol Sin ; 39(3): 422-433, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38499155

RESUMEN

The utilization of enteroviruses engineered with reporter genes serves as a valuable tool for advancing our understanding of enterovirus biology and its applications, enabling the development of effective therapeutic and preventive strategies. In this study, our initial attempts to introduce a NanoLuc luciferase (NLuc) reporter gene into recombinant enteroviruses were unsuccessful in rescuing viable progenies. We hypothesized that the size of the inserted tag might be a determining factor in the rescue of the virus. Therefore, we inserted the 11-amino-acid HiBiT tag into the genomes of enterovirus A71 (EV-A71), coxsackievirus A10 (CVA10), coxsackievirus A7 (CVA7), coxsackievirus A16 (CVA16), namely EV-A71-HiBiT, CVA16-HiBiT, CVA10-HiBiT, CVA7-HiBiT, and observed that the HiBiT-tagged viruses exhibited remarkably high rescue efficiency. Notably, the HiBiT-tagged enteroviruses displayed comparable characteristics to the wild-type viruses. A direct comparison between CVA16-NLuc and CVA16-HiBiT recombinant viruses revealed that the tiny HiBiT insertion had minimal impact on virus infectivity and replication kinetics. Moreover, these HiBiT-tagged enteroviruses demonstrated high genetic stability in different cell lines over multiple passages. In addition, the HiBiT-tagged viruses were successfully tested in antiviral drug assays, and the sensitivity of the viruses to drugs was not affected by the HiBiT tag. Ultimately, our findings provide definitive evidence that the integration of HiBiT into enteroviruses presents a universal, convenient, and invaluable method for advancing research in the realm of enterovirus virology. Furthermore, HiBiT-tagged enteroviruses exhibit great potential for diverse applications, including the development of antivirals and the elucidation of viral infection mechanisms.


Asunto(s)
Enterovirus , Genes Reporteros , Replicación Viral , Enterovirus/genética , Humanos , Luciferasas/genética , Línea Celular , Genoma Viral/genética , Virología/métodos
19.
J Hepatol ; 59(3): 442-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23665181

RESUMEN

BACKGROUND & AIMS: Interferon-γ (IFN-γ), a cytokine produced by activated natural killer cells (NK) and T lymphocytes, is an important regulator of innate and adaptive immunity during hepatitis C virus (HCV) infection. However, the cellular sources and mechanisms of IFN-γ induction in HCV-infection are not fully understood. METHODS: We cultured normal human peripheral blood mononuclear cells (PBMCs) with different populations of immune cells and JFH-1 HCV-infected HuH7.5 (JFH-1/HuH7.5) cells. RESULTS: We found that PBMCs produced large amounts of IFN-γ after co-culture with JFH-1/HuH7.5 cells. Using intracellular cytokine staining, we confirmed that NK cells and NKT cells (to a lesser extent) were the major IFN-γ producers within PBMCs. Purified NK/NKT cells did not produce IFN-γ in response to JFH-1/HuH7.5 cells and depletion of accessory (HLA-DR(+)) cells prevented IFN-γ induction in PBMCs. Through selective cell depletion of dendritic cells or monocytes from PBMCs, we determined that plasmacytoid dendritic cells (pDCs) were indispensable for NK-IFN-γ induction and the presence of monocytes was needed for maximal NK-IFN-γ induction. We further revealed that NK-IFN-γ induction depended on pDC-derived IFN-α while other IFN-γ inducing cytokines, IL-12, and IL-18, played minimal roles. Close contact between JFH-1/HuH7.5 cells and NK cells was required for IFN-γ production and monocyte-derived IL-15 significantly augmented IFN-γ induction. CONCLUSIONS: We discovered a novel mechanism where NK cells interact with pDCs and monocytes, efficiently producing IFN-γ in response to HCV-infected cells. This indicates that co-operation between NK cells and accessory cells is critical for IFN-γ production and regulation of immunity during HCV infection.


Asunto(s)
Hepacivirus/inmunología , Hepacivirus/patogenicidad , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Células Dendríticas/inmunología , Hepatitis C/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Interferón Tipo I/biosíntesis , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/virología , Monocitos/inmunología
20.
Adv Ther ; 40(3): 1171-1186, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36652176

RESUMEN

INTRODUCTION: Patients with chronic hepatitis B (CHB) have a dynamic disease process and risk of end-stage liver disease. It is critical to unambiguously differentiate the stages of the disease and focus on therapy prior to onset of an irreversible clinical endpoint. METHODS: We retrospectively analyzed a wide range of CHB patients at different stages. The predictive power of serum complement component 3 (C3) levels for the development of acute-on-chronic liver failure (ACLF) in patients with decompensated cirrhosis was established and validated. RESULTS: The decrease in serum C3 levels paralleled the severity of diseases related to hepatitis B virus. Patients with decompensated cirrhosis who developed ACLF had significantly lower serum C3 levels than others on admission (0.50 vs. 0.80 g/L, P < 0.001). Data analysis also revealed that low serum C3 was a significant risk factor for developing ACLF (hazard ratio = 0.32, P < 0.01). The area under the receiver operating characteristic curve (auROC) for serum C3 levels that predicted the development of ACLF in patients with decompensated cirrhosis was 0.90, which had sensitivity and specificity of 88.2% and 88.7%, respectively. A similar result was observed in the validation set (auROC = 0.86 for predicting development of ACLF in patients with decompensated cirrhosis). CONCLUSIONS: Serum C3 levels are valuable in assessing the severity of CHB-related stages. Low C3 levels signifies the development of ACLF in patients with decompensated cirrhosis.


Generally, acute-on-chronic liver failure is a rapidly worsening liver failure syndrome. This disease is intractable and with high mortality. Acute decompensation of the liver is defined as the occurrence of complications of liver disease (i.e., ascites, hepatic encephalopathy, gastrointestinal bleeding, and bacterial infection). Clinically, acute decompensation in hepatitis B virus-related cirrhosis (a result of chronic liver injury by virus) often develops into acute-on-chronic liver failure. In addition, the complement component 3 is a serum protein, which participates in the immune response against virus infection and has been reported to be associated with liver failure. We tried to explore the feature of serum complement component 3 to differentiate the stages of the disease and assess its predictive value for acute-on-chronic liver failure. So, we analyzed the complement component 3 data from a broad range of hepatitis B virus-cirrhosis patients. Through analysis, we found that complement component 3 levels are valuable in assessing the severity of chronic hepatitis B-related stages. Low complement component 3 levels can also signify the development of acute-on-chronic liver failure in patients with decompensated cirrhosis.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada , Hepatitis B Crónica , Humanos , Insuficiencia Hepática Crónica Agudizada/complicaciones , Hepatitis B Crónica/complicaciones , Complemento C3 , Cirrosis Hepática/complicaciones , Estudios Retrospectivos , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA