Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 724
Filtrar
1.
Int J Biol Macromol ; 269(Pt 2): 132169, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723801

RESUMEN

In our study, we developed a point of care electrochemical biosensing platform based on the functionalized cysteine-positioned gold electrode to diagnose yellow fever disease from human plasma samples. The developed platform underwent characterization through diverse methods encompassing cyclic voltammetry, electrochemical impedance spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy, and density-functional theory. The capacitive interaction between yellow fever virus non-structural antigen and antibody gave a cathodic signal at approximately -260 mV, and increased in proportion to the amount of non-structural antibody. The created electrochemical biosensor has an ability to detect 96 ag/mL of the yellow fever non-structural antibody with an extensive analytical range varied from 0.1 fg/mL to 1 µg/mL. The interference effects of various substances that could be found in human plasma, and the performance of the method were examined from the point of recovery and relative standard deviation for human plasma samples; hereby, the results confirmed the unprecedented selectivity and accuracy of the proposed method.


Asunto(s)
Técnicas Biosensibles , Técnicas Electroquímicas , Proteínas no Estructurales Virales , Fiebre Amarilla , Humanos , Técnicas Biosensibles/métodos , Fiebre Amarilla/diagnóstico , Fiebre Amarilla/sangre , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/sangre , Técnicas Electroquímicas/métodos , Sistemas de Atención de Punto , Virus de la Fiebre Amarilla/inmunología , Teoría Funcional de la Densidad , Electrodos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Oro/química
2.
PLoS Pathog ; 20(4): e1012167, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38662771

RESUMEN

Dengue virus (DENV) is a medically important flavivirus causing an estimated 50-100 million dengue cases annually, some of whom progress to severe disease. DENV non-structural protein 1 (NS1) is secreted from infected cells and has been implicated as a major driver of dengue pathogenesis by inducing endothelial barrier dysfunction. However, less is known about how DENV NS1 interacts with immune cells and what role these interactions play. Here we report that DENV NS1 can trigger activation of inflammasomes, a family of cytosolic innate immune sensors that respond to infectious and noxious stimuli, in mouse and human macrophages. DENV NS1 induces the release of IL-1ß in a caspase-1 dependent manner. Additionally, we find that DENV NS1-induced inflammasome activation is independent of the NLRP3, Pyrin, and AIM2 inflammasome pathways, but requires CD14. Intriguingly, DENV NS1-induced inflammasome activation does not induce pyroptosis and rapid cell death; instead, macrophages maintain cellular viability while releasing IL-1ß. Lastly, we show that caspase-1/11-deficient, but not NLRP3-deficient, mice are more susceptible to lethal DENV infection. Together, these results indicate that the inflammasome pathway acts as a sensor of DENV NS1 and plays a protective role during infection.


Asunto(s)
Virus del Dengue , Dengue , Inflamasomas , Macrófagos , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Inflamasomas/metabolismo , Inflamasomas/inmunología , Dengue/inmunología , Dengue/virología , Dengue/metabolismo , Ratones , Virus del Dengue/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Interleucina-1beta/metabolismo , Interleucina-1beta/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Caspasa 1/metabolismo
3.
Front Immunol ; 15: 1294898, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38660301

RESUMEN

Human adenovirus type 7 (HAdV-7) is a significant viral pathogen that causes respiratory infections in children. Currently, there are no specific antiviral drugs or vaccines for children targeting HAdV-7, and the mechanisms of its pathogenesis remain unclear. The NLRP3 inflammasome-driven inflammatory cascade plays a crucial role in the host's antiviral immunity. Our previous study demonstrated that HAdV-7 infection activates the NLRP3 inflammasome. Building upon this finding, our current study has identified the L4 100 kDa protein encoded by HAdV-7 as the primary viral component responsible for NLRP3 inflammasome activation. By utilizing techniques such as co-immunoprecipitation, we have confirmed that the 100 kDa protein interacts with the NLRP3 protein and facilitates the assembly of the NLRP3 inflammasome by binding specifically to the NACHT and LRR domains of NLRP3. These insights offer a deeper understanding of HAdV-7 pathogenesis and contribute to the development of novel antiviral therapies.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas no Estructurales Virales , Humanos , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/inmunología , Adenovirus Humanos/fisiología , Células HEK293 , Inflamasomas/metabolismo , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Unión Proteica , Proteínas Virales/metabolismo , Proteínas Virales/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo
4.
Virol Sin ; 39(2): 264-276, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38272236

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is a major economically devastating pathogen that has evolved various strategies to evade innate immunity. Downregulation of antiviral interferon largely promotes PRRSV immunoevasion by utilizing cytoplasmic melanoma differentiation-associated gene 5 (MDA5), a receptor that senses viral RNA. In this study, the downregulated transcription and expression levels of porcine MDA5 in PRRSV infection were observed, and the detailed mechanisms were explored. We found that the interaction between P62 and MDA5 is enhanced due to two factors: the phosphorylation modification of the autophagic receptor P62 by the upregulated kinase CK2α and the K63 ubiquitination of porcine MDA5 catalyzed by the E3 ubiquitinase TRIM21 in PRRSV-infected cells. As a result of these modifications, the classic P62-mediated autophagy is triggered. Additionally, porcine MDA5 interacts with the chaperonin containing TCP1 subunit 2 (CCT2), which is enhanced by PRRSV nsp3. This interaction promotes the aggregate formation and autophagic clearance of MDA5-CCT2-nsp3 independently of ubiquitination. In summary, enhanced MDA5 degradation occurs in PRRSV infection via two autophagic pathways: the binding of MDA5 with the autophagy receptor P62 and the aggrephagy receptor CCT2, leading to intense innate immune suppression. The research reveals a novel mechanism of immune evasion in PRRSV infection and provides fundamental insights for the development of new vaccines or therapeutic strategies.


Asunto(s)
Autofagia , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1 , Virus del Síndrome Respiratorio y Reproductivo Porcino , Animales , Línea Celular , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Fosforilación , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Porcinos , Ubiquitinación , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Humanos
5.
Viruses ; 14(11)2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36366430

RESUMEN

Zika virus (ZIKV)-specific T cells are activated by different peptides derived from virus structural and nonstructural proteins, and contributed to the viral clearance or protective immunity. Herein, we have depicted the profile of CD8+ and CD4+ T cell immunogenicity of ZIKV proteins in C57BL/6 (H-2b) and BALB/c (H-2d) mice, and found that featured cellular immunity antigens were variant among different murine alleles. In H-2b mice, the proteins E, NS2, NS3 and NS5 are recognized as immunodominant antigens by CD8+ T cells, while NS4 is dominantly recognized by CD4+ T cells. In contrast, in H-2d mice, NS1 and NS4 are the dominant CD8+ T cell antigen and NS4 as the dominant CD4+ T cell antigen, respectively. Among the synthesized 364 overlapping polypeptides spanning the whole proteome of ZIKV, we mapped 91 and 39 polypeptides which can induce ZIKV-specific T cell responses in H-2b and H-2d mice, respectively. Through the identification of CD8+ T cell epitopes, we found that immunodominant regions E294-302 and NS42351-2360 are hotspots epitopes with a distinct immunodominance hierarchy present in H-2b and H-2d mice, respectively. Our data characterized an overall landscape of the immunogenic spectrum of the ZIKV polyprotein, and provide useful insight into the vaccine development.


Asunto(s)
Vacunas , Infección por el Virus Zika , Virus Zika , Animales , Ratones , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Epítopos Inmunodominantes , Ratones Endogámicos C57BL , Infección por el Virus Zika/prevención & control , Proteínas no Estructurales Virales/inmunología , Proteínas del Envoltorio Viral/inmunología
6.
J Virol ; 96(18): e0081822, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36098513

RESUMEN

Tick-borne encephalitis virus (TBEV) is an important human arthropod-borne virus that causes tick-borne encephalitis (TBE) in humans. TBEV acutely infects the central nervous system (CNS), leading to neurological symptoms of various severity. No therapeutics are currently available for TBEV-associated disease. Virus strains of various pathogenicity have been described, although the basis of their diverse clinical outcome remains undefined. Work with infectious TBEV requires high-level biocontainment, meaning model systems that can recapitulate the virus life cycle are highly sought. Here, we report the generation of a self-replicating, noninfectious TBEV replicon used to study properties of high (Hypr) and low (Vs) pathogenic TBEV isolates. Using a Spinach2 RNA aptamer and luciferase reporter system, we perform the first direct comparison of Hypr and Vs in cell culture. Infectious wild-type (WT) viruses and chimeras of the nonstructural proteins 3 (NS3) and 5 (NS5) were investigated in parallel to validate the replicon data. We show that Hypr replicates to higher levels than Vs in mammalian cells, but not in arthropod cells, and that the basis of these differences map to the NS5 region, encoding the methyltransferase and RNA polymerase. For both Hypr and Vs strains, NS5 and the viral genome localized to intracellular structures typical of positive-strand RNA viruses. Hypr was associated with significant activation of IRF-3, caspase-3, and caspase-8, while Vs activated Akt, affording protection against caspase-mediated apoptosis. Higher activation of stress-granule proteins TIAR and G3BPI were an additional early feature of Vs but not for Hypr. These findings highlight novel host cell responses driven by NS5 that may dictate the differential clinical characteristics of TBEV strains. This highlights the utility of the TBEV replicons for further virological characterization and antiviral drug screening. IMPORTANCE Tick-borne encephalitis virus (TBEV) is an emerging virus of the flavivirus family that is spread by ticks and causes neurological disease of various severity. No specific therapeutic treatments are available for TBE, and control in areas of endemicity is limited to vaccination. The pathology of TBEV ranges from mild to fatal, depending on the virus genotype. Characterization of TBEV isolates is challenging due to the requirement for high-containment facilities. Here, we described the construction of novel TBEV replicons that permit a molecular comparison of TBEV isolates of high and low pathogenicity.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Interacciones Microbiota-Huesped , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Activación Enzimática , Factor 3 Regulador del Interferón/genética , Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas no Estructurales Virales/inmunología
7.
Cell Rep ; 38(10): 110503, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35235832

RESUMEN

Natural killer (NK) cells are innate immune cells that contribute to host defense against virus infections. NK cells respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and are activated in patients with acute coronavirus disease 2019 (COVID-19). However, by which mechanisms NK cells detect SARS-CoV-2-infected cells remains largely unknown. Here, we show that the Non-structural protein 13 of SARS-CoV-2 encodes for a peptide that is presented by human leukocyte antigen E (HLA-E). In contrast with self-peptides, the viral peptide prevents binding of HLA-E to the inhibitory receptor NKG2A, thereby rendering target cells susceptible to NK cell attack. In line with these observations, NKG2A-expressing NK cells are particularly activated in patients with COVID-19 and proficiently limit SARS-CoV-2 replication in infected lung epithelial cells in vitro. Thus, these data suggest that a viral peptide presented by HLA-E abrogates inhibition of NKG2A+ NK cells, resulting in missing self-recognition.


Asunto(s)
COVID-19 , Antígenos de Histocompatibilidad Clase I , Células Asesinas Naturales , Metiltransferasas , Subfamília C de Receptores Similares a Lectina de Células NK , ARN Helicasas , SARS-CoV-2 , Proteínas no Estructurales Virales , COVID-19/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/inmunología , Metiltransferasas/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Péptidos/metabolismo , ARN Helicasas/inmunología , Proteínas no Estructurales Virales/inmunología , Antígenos HLA-E
8.
Open Biol ; 12(3): 210320, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35232252

RESUMEN

Hepatitis C virus (HCV) remains a global public health challenge with an estimated 71 million people chronically infected, with surges in new cases and no effective vaccine. New methods are needed to study the human immune response to HCV since in vivo animal models are limited and in vitro cancer cell models often show dysregulated immune and proliferative responses. Here, we developed a CD8+ T cell and adult stem cell liver organoid system using a microfluidic chip to coculture 3D human liver organoids embedded in extracellular matrix with HLA-matched primary human T cells in suspension. We then employed automated phase contrast and immunofluorescence imaging to monitor T cell invasion and morphological changes in the liver organoids. This microfluidic coculture system supports targeted killing of liver organoids when pulsed with a peptide specific for HCV non-structural protein 3 (NS3) (KLVALGINAV) in the presence of patient-derived CD8+ T cells specific for KLVALGINAV. This demonstrates the novel potential of the coculture system to molecularly study adaptive immune responses to HCV in an in vitro setting using primary human cells.


Asunto(s)
Linfocitos T CD8-positivos , Hepatitis C , Organoides , Linfocitos T CD8-positivos/inmunología , Técnicas de Cocultivo , Hepacivirus , Hepatitis C/inmunología , Humanos , Microfluídica , Proteínas no Estructurales Virales/inmunología
9.
Nat Commun ; 13(1): 105, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013224

RESUMEN

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Canales Iónicos/genética , Proteínas de la Membrana/genética , Infección por el Virus Zika/genética , Virus Zika/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Línea Celular Tumoral , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Femenino , Regulación de la Expresión Génica , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Aparato de Golgi/virología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Canales Iónicos/deficiencia , Canales Iónicos/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Virus Zika/crecimiento & desarrollo , Virus Zika/patogenicidad , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
10.
Curr Mol Med ; 22(1): 50-66, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33622224

RESUMEN

The proteins of coronavirus are classified as non-structural, structural, and accessory. There are 16 non-structural viral proteins besides their precursors (1a and 1ab polyproteins). The non-structural proteins are named nsp1 to nsp16, and they act as enzymes, coenzymes, and binding proteins to facilitate the replication, transcription, and translation of the virus. The structural proteins are bound to the RNA in the nucleocapsid (N- protein) or to the lipid bilayer membrane of the viral envelope. The lipid bilayer proteins include the membrane protein (M), an envelope protein (E), and spike protein (S). Besides their role as structural proteins, they are essential for the host cells' binding and invasion. The SARS-CoV-2 contains six accessory proteins which participate in the viral replication, assembly and virus-host interactions. The SARS-CoV-2 accessory proteins are orf3a, orf6, orf7a, orf7b, orf8, and orf10. The functions of the SARS-CoV-2 are not well known, while the functions of their corresponding proteins in SARS-CoV are either well known or poorly studied. Recently, the Oxford University and Astrazeneca, Pfizer and BioNTech have made SARS-CoV-2 vaccines by targeting the spike protein gene. The US Food and Drug Administration (FDA) and the health authorities of the United Kingdom have approved and started conducting vaccinations using the Pfizer and BioNTech mRNA vaccine. Also, The FDA of the USA has approved the use of two monoclonal antibodies produced by Regeneron pharmaceuticals to target the spike protein for treating COVID-19. The SARS-CoV-2 proteins can be used for the diagnosis, as drug targets and in vaccination trials for COVID-19. In future COVID-19 research, more efforts should be made to elaborate the functions and structure of the SARS-CoV- 2 proteins so as to use them as targets for COVID-19 drugs and vaccines. Special attention should be paid to extensive research on the SARS-CoV-2 nsp3, orf8, and orf10.


Asunto(s)
Antivirales/farmacología , Vacunas contra la COVID-19 , COVID-19/prevención & control , SARS-CoV-2/química , Proteínas Virales/efectos de los fármacos , Proteínas Virales/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/inmunología , COVID-19/inmunología , Diseño de Fármacos , Humanos , Inmunoterapia , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Desarrollo de Vacunas , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/fisiología , Proteínas Virales/fisiología , Proteínas Reguladoras y Accesorias Virales/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/inmunología , Proteínas Reguladoras y Accesorias Virales/fisiología , Proteínas Estructurales Virales/efectos de los fármacos , Proteínas Estructurales Virales/inmunología , Proteínas Estructurales Virales/fisiología , Vacunas de ARNm , Tratamiento Farmacológico de COVID-19
11.
J Virol ; 96(3): e0161421, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34787454

RESUMEN

Bluetongue, caused by bluetongue virus (BTV), is a widespread arthropod-borne disease of ruminants that entails a recurrent threat to the primary sector of developed and developing countries. In this work, we report modified vaccinia virus Ankara (MVA) and ChAdOx1-vectored vaccines designed to simultaneously express the immunogenic NS1 protein and/or NS2-Nt, the N-terminal half of protein NS2 (NS21-180). A single dose of MVA or ChAdOx1 expressing NS1-NS2-Nt improved the protection conferred by NS1 alone in IFNAR(-/-) mice. Moreover, mice immunized with ChAdOx1/MVA-NS1, ChAdOx1/MVA-NS2-Nt, or ChAdOx1/MVA-NS1-NS2-Nt developed strong cytotoxic CD8+ T-cell responses against NS1, NS2-Nt, or both proteins and were fully protected against a lethal infection with BTV serotypes 1, 4, and 8. Furthermore, although a single immunization with ChAdOx1-NS1-NS2-Nt partially protected sheep against BTV-4, the administration of a booster dose of MVA-NS1-NS2-Nt promoted a faster viral clearance, reduction of the period and level of viremia and also protected from the pathology produced by BTV infection. IMPORTANCE Current BTV vaccines are effective but they do not allow to distinguish between vaccinated and infected animals (DIVA strategy) and are serotype specific. In this work we have develop a DIVA multiserotype vaccination strategy based on adenoviral (ChAdOx1) and MVA vaccine vectors, the most widely used in current phase I and II clinical trials, and the conserved nonstructural BTV proteins NS1 and NS2. This immunization strategy solves the major drawbacks of the current marketed vaccines.


Asunto(s)
Virus de la Lengua Azul/inmunología , Lengua Azul/prevención & control , Vectores Genéticos/genética , Virus Vaccinia/genética , Proteínas no Estructurales Virales/genética , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Lengua Azul/clasificación , Vectores Genéticos/inmunología , Inmunidad Celular , Inmunización , Inmunogenicidad Vacunal , Serogrupo , Ovinos , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Virus Vaccinia/inmunología , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
12.
Mol Immunol ; 142: 11-21, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34959069

RESUMEN

Senecavirus A (SVA) is an oncolytic virus, which can propagate in human tumor cells and has been used as an oncolytic virotherapy candidate in humans. Besides, SVA circulates in pigs and causes vesicles and coalescing erosions on the snouts and coronary bands in infected pigs and results in neonatal morbidity. SVA has evolved the ability to suppress host innate immune response to benefit viral replication. SVA 3Cpro and 2C protein inhibit the production of host type I interferon (IFN) by degradation of several components of innate immune pathway. In this study, for the first time, we determined that SVA 2B antagonized host innate immune response in both human and porcine cells. SVA 2B protein degraded mitochondrial antiviral-signaling protein (MAVS), a key host molecule in the innate immune pathway, and a colocalization and interaction between 2B and MAVS was observed in the context of viral infection. Further study showed that the 1-48 and 100-128 regions of 2B were essential for inhibition of type I IFN expression. In addition, we determined that 2B degraded MAVS depending on caspase-9 and caspase-3. In conclusion, our results revealed a novel strategy for SVA 2B protein to antagonize host innate immune response, which will help for clarification of the pathogenesis of SVA and provide an insight for oncolytic virotherapy of SVA.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Picornaviridae/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/inmunología , Viroterapia Oncolítica/métodos , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Porcinos , Proteínas no Estructurales Virales/genética , Proteínas Viroporinas/genética , Proteínas Viroporinas/inmunología , Liberación del Virus/fisiología , Replicación Viral/fisiología
13.
Viruses ; 13(11)2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34835021

RESUMEN

It is generally believed that a successful Zika virus (ZIKV) vaccine should induce neutralizing antibodies against the ZIKV envelope (E) protein to efficiently halt viral infection. However, E-specific neutralizing antibodies have been implicated in a phenomenon called antibody-dependent enhancement, which represents an ongoing concern in the flavivirus-vaccinology field. In this report, we investigated the vaccination potential of replication-deficient adenoviral vectors encoding the ZIKV non-structural proteins 1 and 2 (NS1/NS2) and employed the strategy of linking the antigens to the MHC-II associated invariant chain (li) to improve immunogenicity and by inference, the level of protection. We demonstrated that li-linkage enhanced the production of anti-NS1 antibodies and induced an accelerated and prolonged polyfunctional CD8 T cell response in mice, which ultimately resulted in a high degree of protection against ZIKV infection of the CNS.


Asunto(s)
Antígenos Virales/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/prevención & control , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Femenino , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos C57BL , Vacunación , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/inmunología , Virus Zika/inmunología , Infección por el Virus Zika/virología
14.
Biomed Pharmacother ; 144: 112304, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34634560

RESUMEN

Dengue virus (DENV) is a global health threat causing about half of the worldwide population to be at risk of infection, especially the people living in tropical and subtropical area. Although the dengue disease caused by dengue virus (DENV) is asymptomatic and self-limiting in most people with first infection, increased severe dengue symptoms may be observed in people with heterotypic secondary DENV infection. Since there is a lack of specific antiviral medication, the development of dengue vaccines is critical in the prevention and control this disease. Several targets and strategies in the development of dengue vaccine have been demonstrated. Currently, Dengvaxia, a live-attenuated chimeric yellow-fever/tetravalent dengue vaccine (CYD-TDV) developed by Sanofi Pasteur, has been licensed and approved for clinical use in some countries. However, this vaccine has demonstrated low efficacy in children and dengue-naïve individuals and also increases the risk of severe dengue in young vaccinated recipients. Accordingly, many novel strategies for the dengue vaccine are under investigation and development. Here, we conducted a systemic literature review according to PRISMA guidelines to give a concise overview of various aspects of the vaccine development process against DENVs, mainly targeting five potential strategies including live attenuated vaccine, inactivated virus vaccine, recombinant subunit vaccine, viral-vector vaccine, and DNA vaccine. This study offers the comprehensive view of updated information and current progression of immunogen selection as well as strategies of vaccine development against DENVs.


Asunto(s)
Vacunas contra el Dengue/uso terapéutico , Virus del Dengue/inmunología , Dengue/prevención & control , Desarrollo de Vacunas , Proteínas del Envoltorio Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Dengue/inmunología , Dengue/virología , Vacunas contra el Dengue/efectos adversos , Vacunas contra el Dengue/inmunología , Virus del Dengue/genética , Virus del Dengue/patogenicidad , Humanos , Resultado del Tratamiento , Eficacia de las Vacunas , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/uso terapéutico , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico , Proteínas del Envoltorio Viral/genética , Proteínas no Estructurales Virales/genética
15.
J Virol ; 95(20): e0023121, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34379499

RESUMEN

The NS1 protein of the influenza A virus plays a critical role in regulating several biological processes in cells, including the type I interferon (IFN) response. We previously profiled the cellular factors that interact with the NS1 protein of influenza A virus and found that the NS1 protein interacts with proteins involved in RNA splicing/processing, cell cycle regulation, and protein targeting processes, including 14-3-3ε. Since 14-3-3ε plays an important role in retinoic acid-inducible gene I (RIG-I) translocation to mitochondrial antiviral-signaling protein (MAVS) to activate type I IFN expression, the interaction of the NS1 and 14-3-3ε proteins may prevent the RIG-I-mediated IFN response. In this study, we confirmed that the 14-3-3ε protein interacts with the N-terminal domain of the NS1 protein and that the NS1 protein inhibits RIG-I-mediated IFN-ß promoter activation in 14-3-3ε-overexpressing cells. In addition, our results showed that knocking down 14-3-3ε can reduce IFN-ß expression elicited by influenza A virus and enhance viral replication. Furthermore, we found that threonine in the 49th amino acid position of the NS1 protein plays a role in the interaction with 14-3-3ε. Influenza A virus expressing C terminus-truncated NS1 with a T49A mutation dramatically increases IFN-ß mRNA in infected cells and causes slower replication than that of virus without the T-to-A mutation. Collectively, this study demonstrates that 14-3-3ε is involved in influenza A virus-initiated IFN-ß expression and that the interaction of the NS1 protein and 14-3-3ε may be one of the mechanisms for inhibiting type I IFN activation during influenza A virus infection. IMPORTANCE Influenza A virus is an important human pathogen causing severe respiratory disease. The virus has evolved several strategies to dysregulate the innate immune response and facilitate its replication. We demonstrate that the NS1 protein of influenza A virus interacts with the cellular chaperone protein 14-3-3ε, which plays a critical role in retinoic acid-inducible gene I (RIG-I) translocation that induces type I interferon (IFN) expression, and that NS1 protein prevents RIG-I translocation to the mitochondrial membrane. The interaction site for 14-3-3ε is the RNA-binding domain (RBD) of the NS1 protein. Therefore, this research elucidates a novel mechanism by which the NS1 RBD mediates IFN-ß suppression to facilitate influenza A viral replication. Additionally, the findings reveal the antiviral role of 14-3-3ε during influenza A virus infection.


Asunto(s)
Proteínas 14-3-3/inmunología , Gripe Humana/inmunología , Interferón beta/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular Tumoral , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/fisiología , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , ARN Viral/genética , Receptores Inmunológicos/metabolismo , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
16.
Signal Transduct Target Ther ; 6(1): 304, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34404759

RESUMEN

A comprehensive analysis of the humoral immune response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is essential in understanding COVID-19 pathogenesis and developing antibody-based diagnostics and therapy. In this work, we performed a longitudinal analysis of antibody responses to SARS-CoV-2 proteins in 104 serum samples from 49 critical COVID-19 patients using a peptide-based SARS-CoV-2 proteome microarray. Our data show that the binding epitopes of IgM and IgG antibodies differ across SARS-CoV-2 proteins and even within the same protein. Moreover, most IgM and IgG epitopes are located within nonstructural proteins (nsps), which are critical in inactivating the host's innate immune response and enabling SARS-CoV-2 replication, transcription, and polyprotein processing. IgM antibodies are associated with a good prognosis and target nsp3 and nsp5 proteases, whereas IgG antibodies are associated with high mortality and target structural proteins (Nucleocapsid, Spike, ORF3a). The epitopes targeted by antibodies in patients with a high mortality rate were further validated using an independent serum cohort (n = 56) and using global correlation mapping analysis with the clinical variables that are associated with COVID-19 severity. Our data provide fundamental insight into humoral immunity during SARS-CoV-2 infection. SARS-CoV-2 immunogenic epitopes identified in this work could also help direct antibody-based COVID-19 treatment and triage patients.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Inmunidad Humoral , SARS-CoV-2/inmunología , Proteínas no Estructurales Virales/inmunología , COVID-19/mortalidad , Enfermedad Crítica , Supervivencia sin Enfermedad , Epítopos/inmunología , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , Análisis por Matrices de Proteínas , Tasa de Supervivencia
17.
Int J Mol Sci ; 22(13)2021 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-34206987

RESUMEN

Hepatitis C virus (HCV) is one of the main triggers of chronic liver disease. Despite tremendous progress in the HCV field, there is still no vaccine against this virus. Potential vaccines can be based on its recombinant proteins. To increase the humoral and, especially, cellular immune response to them, more effective adjuvants are needed. Here, we evaluated a panel of compounds as potential adjuvants using the HCV NS5B protein as an immunogen. These compounds included inhibitors of polyamine biosynthesis and urea cycle, the mTOR pathway, antioxidants, and cellular receptors. A pronounced stimulation of cell proliferation and interferon-γ (IFN-γ) secretion in response to concanavalin A was shown for antioxidant N-acetylcysteine (NAC), polyamine biosynthesis inhibitor 2-difluoromethylornithine (DFMO), and TLR9 agonist CpG ODN 1826 (CpG). Their usage during the immunization of mice with the recombinant NS5B protein significantly increased antibody titers, enhanced lymphocyte proliferation and IFN-γ production. NAC and CpG decreased relative Treg numbers; CpG increased the number of myeloid-derived suppressor cells (MDSCs), whereas neither NAC nor DFMO affected MDSC counts. NAC and DFMO suppressed NO and interleukin 10 (IL-10) production by splenocytes, while DFMO increased the levels of IL-12. This is the first evidence of immunomodulatory activity of NAC and DFMO during prophylactic immunization against infectious diseases.


Asunto(s)
Acetilcisteína/farmacología , Adyuvantes Inmunológicos/farmacología , Eflornitina/farmacología , Hepatitis C/inmunología , Inmunidad Activa/efectos de los fármacos , Proteínas no Estructurales Virales/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Inmunogenicidad Vacunal/efectos de los fármacos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos DBA , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Óxido Nítrico/metabolismo , Oligodesoxirribonucleótidos/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Vacunas contra Hepatitis Viral/inmunología
18.
PLoS One ; 16(7): e0253364, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34270554

RESUMEN

Of the 16 non-structural proteins (Nsps) encoded by SARS CoV-2, Nsp3 is the largest and plays important roles in the viral life cycle. Being a large, multidomain, transmembrane protein, Nsp3 has been the most challenging Nsp to characterize. Encoded within Nsp3 is the papain-like protease domain (PLpro) that cleaves not only the viral polypeptide but also K48-linked polyubiquitin and the ubiquitin-like modifier, ISG15, from host cell proteins. We here compare the interactors of PLpro and Nsp3 and find a largely overlapping interactome. Intriguingly, we find that near full length Nsp3 is a more active protease compared to the minimal catalytic domain of PLpro. Using a MALDI-TOF based assay, we screen 1971 approved clinical compounds and identify five compounds that inhibit PLpro with IC50s in the low micromolar range but showed cross reactivity with other human deubiquitinases and had no significant antiviral activity in cellular SARS-CoV-2 infection assays. We therefore looked for alternative methods to block PLpro activity and engineered competitive nanobodies that bind to PLpro at the substrate binding site with nanomolar affinity thus inhibiting the enzyme. Our work highlights the importance of studying Nsp3 and provides tools and valuable insights to investigate Nsp3 biology during the viral infection cycle.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteasas/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Anticuerpos de Cadena Única/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Células A549 , Complejo Antígeno-Anticuerpo , Humanos , Concentración 50 Inhibidora , ARN Polimerasa Dependiente del ARN/inmunología , ARN Polimerasa Dependiente del ARN/metabolismo , Anticuerpos de Cadena Única/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo
19.
J Virol ; 95(19): e0061921, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34232731

RESUMEN

Although transmission of Zika virus (ZIKV) in the Americas has greatly declined since late 2017, recent reports of reduced risks of symptomatic Zika by prior dengue virus (DENV) infection and increased risks of severe dengue disease by previous ZIKV or DENV infection underscore a critical need for serological tests that can discriminate past ZIKV, DENV, and/or other flavivirus infections and improve our understanding of the immune interactions between these viruses and vaccine strategy in endemic regions. As serological tests for ZIKV primarily focus on envelope (E) and nonstructural protein 1 (NS1), antibodies to other ZIKV proteins have not been explored. Here, we employed Western blot analysis using antigens of 6 flaviviruses from 3 serocomplexes to investigate antibody responses following reverse transcription-PCR (RT-PCR)-confirmed ZIKV infection. Panels of 20 primary ZIKV and 20 ZIKV with previous DENV infection recognized E proteins of all 6 flaviviruses and the NS1 protein of ZIKV with some cross-reactivity to DENV. While the primary ZIKV panel recognized only the premembrane (prM) protein of ZIKV, the ZIKV with previous DENV panel recognized both ZIKV and DENV prM proteins. Analysis of antibody responses following 42 DENV and 18 West Nile virus infections revealed similar patterns of recognition by anti-E and anti-NS1 antibodies, whereas both panels recognized the prM protein of the homologous serocomplex but not others. The specificity was further supported by analysis of sequential samples. Together, these findings suggest that anti-prM antibody is a flavivirus serocomplex-specific marker and can be used to delineate current and past flavivirus infections in endemic areas. IMPORTANCE Despite a decline in Zika virus (ZIKV) transmission since late 2017, questions regarding its surveillance, potential reemergence, and interactions with other flaviviruses in regions where it is endemic remain unanswered. Recent studies have reported reduced risks of symptomatic Zika by prior dengue virus (DENV) infection and increased risks of severe dengue disease by previous ZIKV or DENV infection, highlighting a need for better serological tests to discriminate past ZIKV, DENV, and/or other flavivirus infections and improved understanding of the immune interactions and vaccine strategy for these viruses. As most serological tests for ZIKV focused on envelope and nonstructural protein 1, antibodies to other ZIKV proteins, including potentially specific antibodies, remain understudied. We employed Western blot analysis using antigens of 6 flaviviruses to study antibody responses following well-documented ZIKV, DENV, and West Nile virus infections and identified anti-premembrane antibody as a flavivirus serocomplex-specific marker to delineate current and past flavivirus infections in areas where flaviviruses are endemic.


Asunto(s)
Anticuerpos Antivirales/sangre , Dengue/inmunología , Proteínas del Envoltorio Viral/inmunología , Fiebre del Nilo Occidental/inmunología , Infección por el Virus Zika/inmunología , Anticuerpos Antivirales/inmunología , Western Blotting , Reacciones Cruzadas , Dengue/diagnóstico , Virus del Dengue/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Sensibilidad y Especificidad , Proteínas no Estructurales Virales/inmunología , Fiebre del Nilo Occidental/diagnóstico , Virus del Nilo Occidental/inmunología , Virus Zika/inmunología , Infección por el Virus Zika/diagnóstico
20.
Mol Immunol ; 138: 20-30, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34332182

RESUMEN

Respiratory syncytial virus (RSV) infection in early life is associated strongly with the subsequent development and exacerbation of asthma, however, the mechanism is still ambiguous. In this study, we identified that RSV nonstructural protein (NS) 1 plays a critical role. Plasmid-mediated overexpression of NS1 induced significant airway hyperresponsiveness, eosinophilia, and mucus hyperproduction in mice. In the pNS1 group, there were markedly elevated proportions of Th2 and Th17 cells, while Th1 and Foxp3+ regulatory T cells (Tregs) significantly declined compared with the control group. Serum concentrations of interleukin (IL)-4, IL-5, IL-6, IL-17, transforming growth factor-beta, and tumor necrosis factor-alpha increased but levels of interferon-gamma and interleukin-10 declined in pNS1 group. Besides, NS1 caused a significant rise of serum thymic stromal lymphopoietin (TSLP) and OX40L levels, and a neutralizing mAb anti-OX40L was capable of promoting RSV clearance and attenuating the airway allergic inflammation caused by pNS1. Otherwise, OX40L-blocking counteracts the inhibitory effect of pNS1 on Tregs in the spleen. RSV NS1 caused elevated levels of phospho-AKT, phospho-mTOR, and phospho-S6K1, which were partially attenuated by anti-OX40L. Moreover, a specific inhibitor of mTORC1 significantly relieved the inhibition of Foxp3 expression and Tregs differentiation. Together, the data indicate that RSV NS1 protein breaks immune tolerance and induces airway inflammation and hyperresponsiveness in mice. In this process, NS1-stimulated TSLP and OX40L play a major role by inhibiting the induction of Tregs, which is at least partially mediated by modulating AKT-mTOR signaling pathways.


Asunto(s)
Tolerancia Inmunológica/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Citocinas/inmunología , Regulación hacia Abajo , Femenino , Ratones , Ratones Endogámicos BALB C , Ligando OX40/inmunología , Receptores OX40/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitial Respiratorio Humano/inmunología , Serina-Treonina Quinasas TOR/inmunología , Linfopoyetina del Estroma Tímico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA