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












Base de datos
Intervalo de año de publicación
1.
Emerg Microbes Infect ; 13(1): 2348528, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38662785

RESUMEN

Zika is a systemic inflammatory disease caused by infection with Zika virus (ZIKV). ZIKV infection in adults is associated with encephalitis marked by elevated expression of pro-inflammatory cytokines and chemokines, as well as increased brain infiltration of immune cells. In this study, we demonstrate that ZIKV encephalitis in a mouse infection model exhibits increased brain TSPO expression. TSPO expression on brain-resident and infiltrating immune cells in ZIKV infection correlates with disease and inflammation status in the brain. Brain TSPO expression can also be sensitively detected ex vivo and in vitro using radioactive small molecule probes that specifically bind to TSPO, such as [3H]PK11195. TSPO expression on brain-resident and infiltrating immune cells is a biomarker of ZIKV neuroinflammation, which can also be a general biomarker of acute viral neuroinflammatory disease.


Asunto(s)
Biomarcadores , Encéfalo , Enfermedades Neuroinflamatorias , Receptores de GABA , Infección por el Virus Zika , Virus Zika , Animales , Infección por el Virus Zika/virología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/metabolismo , Ratones , Receptores de GABA/metabolismo , Receptores de GABA/genética , Virus Zika/inmunología , Encéfalo/virología , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Femenino , Citocinas/metabolismo
2.
J Transl Med ; 22(1): 126, 2024 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-38308299

RESUMEN

Glioblastoma multiforme (GBM) is the most common malignant primary brain cancer affecting the adult population. Median overall survival for GBM patients is poor (15 months), primarily due to high rates of tumour recurrence and the paucity of treatment options. Oncolytic virotherapy is a promising treatment alternative for GBM patients, where engineered viruses selectively infect and eradicate cancer cells by inducing cell lysis and eliciting robust anti-tumour immune response. In this study, we evaluated the oncolytic potency of live-attenuated vaccine strains of Zika virus (ZIKV-LAV) against human GBM cells in vitro. Our findings revealed that Axl and integrin αvß5 function as cellular receptors mediating ZIKV-LAV infection in GBM cells. ZIKV-LAV strains productively infected and lysed human GBM cells but not primary endothelia and terminally differentiated neurons. Upon infection, ZIKV-LAV mediated GBM cell death via apoptosis and pyroptosis. This is the first in-depth molecular dissection of how oncolytic ZIKV infects and induces death in tumour cells.


Asunto(s)
Glioblastoma , Viroterapia Oncolítica , Virus Oncolíticos , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/fisiología , Infección por el Virus Zika/prevención & control , Glioblastoma/terapia , Vacunas Atenuadas , Recurrencia Local de Neoplasia/terapia
3.
Eur J Nucl Med Mol Imaging ; 50(3): 742-755, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36348095

RESUMEN

INTRODUCTION: Zika virus (ZIKV) is a neurotropic human pathogen that causes neuroinflammation, whose hallmark is elevated translocator protein (TSPO) expression in the brain. This study investigates ZIKV-associated changes in adult brain TSPO expression, evaluates the effectiveness of TSPO radioligands in detecting TSPO expression, and identifies cells that drive brain TSPO expression in a mouse infection model. METHODS: The interferon-deficient AG129 mouse infected with ZIKV was used as neuroinflammation model. TSPO expression was evaluated by tissue immunostaining. TSPO radioligands, [3H]PK11195 and [18F]FEPPA, were used for in vitro and ex vivo detection of TSPO in infected brains. [18F]FEPPA-PET was used for in vivo detection of TSPO expression. Cell subsets that contribute to TSPO expression were identified by flow cytometry. RESULTS: Brain TSPO expression increased with ZIKV disease severity. This increase was contributed by TSPO-positive microglia and infiltrating monocytes; and by influx of TSPO-expressing immune cells into the brain. [3H]PK11195 and [18F]FEPPA distinguish ZIKV-infected brains from normal controls in vitro and ex vivo. [18F]FEPPA brain uptake by PET imaging correlated with disease severity and neuroinflammation. However, TSPO expression by immune cells contributed to significant blood pool [18F]FEPPA activity which could confound [18F]FEPPA-PET imaging results. CONCLUSIONS: TSPO is a biologically relevant imaging target for ZIKV neuroinflammation. Brain [18F]FEPPA uptake can be a surrogate marker for ZIKV disease and may be a potential PET imaging marker for ZIKV-induced neuroinflammation. Future TSPO-PET/SPECT studies on viral neuroinflammation and related encephalitis should assess the contribution of immune cells on TSPO expression and employ appropriate image correction methods to subtract blood pool activity.


Asunto(s)
Encefalitis , Infección por el Virus Zika , Virus Zika , Adulto , Humanos , Ratones , Animales , Virus Zika/metabolismo , Infección por el Virus Zika/diagnóstico por imagen , Infección por el Virus Zika/metabolismo , Enfermedades Neuroinflamatorias , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Tomografía de Emisión de Positrones/métodos , Modelos Animales de Enfermedad , Receptores de GABA/metabolismo
4.
Eur J Nucl Med Mol Imaging ; 49(13): 4516-4528, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35876869

RESUMEN

PURPOSE: Zika (ZIKV) is a viral inflammatory disease affecting adults, children, and developing fetuses. It is endemic to tropical and sub-tropical countries, resulting in half the global population at risk of infection. Despite this, there are no approved therapies or vaccines against ZIKV disease. Non-invasive imaging biomarkers are potentially valuable tools for studying viral pathogenesis, prognosticating host response to disease, and evaluating in vivo efficacy of experimental therapeutic interventions. In this study, we evaluated [18F]fluorodeoxyglucose ([18F]FDG)-positron emission tomography (PET) as an imaging biomarker of ZIKV disease in a mouse model and correlated metabolic tracer tissue uptake with real-time biochemical, virological, and inflammatory features of tissue infection. METHODS: [18F]FDG-PET/CT imaging was performed in an acute, lethal ZIKV mouse infection model, at increasing stages of disease severity. [18F]FDG-PET findings were corroborated with ex vivo wholemount-tissue autoradiography and tracer biodistribution studies. Tracer uptake was also correlated with in situ tissue disease status, including viral burden and inflammatory response. Immune profiling of the spleen by flow cytometry was performed to identify the immune cell subsets driving tissue pathology and enhancing tracer uptake in ZIKV disease. RESULTS: Foci of increased [18F]FDG uptake were consistently detected in lymphoid tissues-particularly the spleen-of ZIKV-infected animals. Splenic uptake increased with disease severity, and corroborated findings in tissue pathology. Increased splenic uptake also correlated with increased viral replication and elevated expression of pro-inflammatory cytokines within these tissues. ZIKV-infected spleens were characterized by increased infiltration of myeloid cells, as well as increased proliferation of both myeloid and lymphoid cells. The increased cell proliferation correlated with increased tracer uptake in the spleen. Our findings support the use of [18F]FDG as an imaging biomarker to detect and track ZIKV disease in real time and highlight the dependency of affected tissue on the nature of the viral infection. CONCLUSION: [18F]FDG uptake in the spleen is a useful surrogate for interrogating in situ tissue viral burden and inflammation status in this ZIKV murine model.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Ratones , Infección por el Virus Zika/diagnóstico por imagen , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/patología , Virus Zika/metabolismo , Fluorodesoxiglucosa F18/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Distribución Tisular , Tomografía Computarizada por Rayos X , Tomografía de Emisión de Positrones , Tejido Linfoide/metabolismo , Tejido Linfoide/patología , Inflamación/diagnóstico por imagen , Inflamación/metabolismo , Modelos Animales de Enfermedad , Biomarcadores/metabolismo , Citocinas
5.
Int J Mol Sci ; 21(23)2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33291644

RESUMEN

Current methods to detect and monitor pathogens in biological systems are largely limited by the tradeoffs between spatial context and temporal detail. A new generation of molecular tracking that provides both information simultaneously involves in situ detection coupled with non-invasive imaging. An example is antisense imaging that uses antisense oligonucleotide probes complementary to a target nucleotide sequence. In this study, we explored the potential of repurposing antisense oligonucleotides initially developed as antiviral therapeutics as molecular probes for imaging of viral infections in vitro and in vivo. We employed nuclease-resistant phosphorodiamidate synthetic oligonucleotides conjugated with cell-penetrating peptides (i.e., PPMOs) previously established as antivirals for dengue virus serotype-2 (DENV2). As proof of concept, and before further development for preclinical testing, we evaluated its validity as in situ molecular imaging probe for tracking cellular DENV2 infection using live-cell fluorescence imaging. Although the PPMO was designed to specifically target the DENV2 genome, it was unsuitable as in situ molecular imaging probe. This study details our evaluation of the PPMOs to assess specific and sensitive molecular imaging of DENV2 infection and tells a cautionary tale for those exploring antisense oligonucleotides as probes for non-invasive imaging and monitoring of pathogen infections in experimental animal models.


Asunto(s)
Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Hibridación in Situ , Imagen Molecular , Morfolinos/química , Péptidos/química , Replicación Viral/efectos de los fármacos , Animales , Chlorocebus aethiops , Humanos , Ratones , Oligonucleótidos Antisentido , Células Vero
7.
Virol Sin ; 34(3): 262-269, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31016480

RESUMEN

Our previous work has shown that Saffold virus (SAFV) induced several rodent and primate cell lines to undergo apoptosis (Xu et al. in Emerg Microb Infect 3:1-8, 2014), but the essential viral proteins of SAFV involved in apoptotic activity lack study. In this study, we individually transfected the viral proteins of SAFV into HEp-2 and Vero cells to assess their ability to induce apoptosis, and found that the 2B and 3C proteins are proapoptotic. Further investigation indicated the transmembrane domain of the 2B protein is essential for the apoptotic activity and tetramer formation of the 2B protein. Our research provides clues for the possible mechanisms of apoptosis induced by SAFV in different cell lines. It also opens up new directions to study viral proteins (the 2B, 3C protein), and sets the stage for future exploration of any possible link between SAFV, inclusive of its related uncultivable genotypes, and multiple sclerosis.


Asunto(s)
Apoptosis , Picornaviridae/fisiología , Proteínas Virales/genética , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Picornaviridae/genética , Transfección , Células Vero
8.
Emerg Microbes Infect ; 5(10): e109, 2016 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-27729641

RESUMEN

The Saffold virus (SAFV) genome is translated as a single long polyprotein precursor and co-translationally cleaved to yield 12 separate viral proteins. Little is known about the activities of SAFV proteins although their homologs in other picornaviruses have already been described. To further support research on functions and activities of respective viral proteins, we investigated the spatio-temporal distribution of SAFV proteins in Vero and HEp-2 cells that had been either transfected with plasmids that express individual viral proteins or infected with live SAFV. Our results revealed that, with the exception of the Leader (L) protein, all viral proteins were localized in the cytoplasm at all the time points assayed. The L protein was found in the cytoplasm at an early time point but was subsequently translocated to the nucleus of HEp-2, but not Vero, cells. This was observed in both transfected and infected cells. Further mutational analysis of L protein revealed that Threonine 58 of the Ser/Thr-rich domain of L protein is crucial for protein trafficking between the cytoplasm and nucleus in HEp-2 cells. These findings contribute to a deeper understanding and stimulate investigation of the differetial cellular responses of HEp-2 cells in comparison to other mammalian cell lines during SAFV infection.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Theilovirus/genética , Theilovirus/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Citoplasma/virología , Técnica del Anticuerpo Fluorescente , Genoma Viral , Humanos , Mutación , Transporte de Proteínas , Transfección , Células Vero , Proteínas Virales/genética , Proteínas Virales/inmunología , Virión
9.
Sci Rep ; 6: 28876, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27357918

RESUMEN

Enterovirus 71 (EV-A71) is a neurotropic virus that sporadically causes fatal neurologic illness among infected children. Animal models of EV-A71 infection exist, but they do not recapitulate in animals the spectrum of disease and pathology observed in fatal human cases. Specifically, neurogenic pulmonary oedema (NPE)-the main cause of EV-A71 infection-related mortality-is not observed in any of these models. This limits their utility in understanding viral pathogenesis of neurologic infections. We report the development of a mouse model of EV-A71 infection displaying NPE in severely affected animals. We inoculated one-week-old BALB/c mice with an adapted EV-A71 strain and identified clinical signs consistent with observations in human cases and other animal models. We also observed respiratory distress in some mice. At necropsy, we found their lungs to be heavier and incompletely collapsed compared to other mice. Serum levels of catecholamines and histopathology of lung and brain tissues of these mice strongly indicated onset of NPE. The localization of virally-induced brain lesions also suggested a potential pathogenic mechanism for EV-A71-induced NPE. This novel mouse model of virally-induced NPE represents a valuable resource for studying viral mechanisms of neuro-pathogenesis and pre-clinical testing of potential therapeutics and prophylactics against EV-A71-related neurologic complications.


Asunto(s)
Enterovirus Humano A/fisiología , Infecciones por Enterovirus/patología , Edema Pulmonar/patología , Animales , Anticuerpos Neutralizantes/sangre , Encéfalo/metabolismo , Encéfalo/patología , Catecolaminas/metabolismo , Modelos Animales de Enfermedad , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/mortalidad , Infecciones por Enterovirus/virología , Humanos , Estimación de Kaplan-Meier , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Edema Pulmonar/metabolismo , Edema Pulmonar/mortalidad , Edema Pulmonar/virología , Índice de Severidad de la Enfermedad
10.
Emerg Microbes Infect ; 5: e60, 2016 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-27329847

RESUMEN

Enterovirus 71 (EV71) is a neurotrophic virus that causes hand, foot and mouth disease (HFMD) and occasional neurological infection among children. It infects primate cells but not rodent cells, primarily due to the incompatibility between the virus and the expressed form of its receptor, scavenger receptor class B member 2 (SCARB2) protein, on rodent cells (mSCARB2). We previously generated adapted strains (EV71:TLLm and EV71:TLLmv) that were shown to productively infect primate and rodent cell lines and whose genomes exhibited a multitude of non-synonymous mutations compared with the EV71:BS parental virus. In this study, we aimed to identify mutations that are necessary for productive infection of murine cells by EV71:BS. Using reverse genetics and site-directed mutagenesis, we constructed EV71 infectious clones with specific mutations that generated amino acid substitutions in the capsid VP1 and VP2 proteins. We subsequently assessed the infection induced by clone-derived viruses (CDVs) in mouse embryonic fibroblast NIH/3T3 and murine neuroblastoma Neuro-2a cell lines. We found that the CDV:BS-VP1(K98E,E145A,L169F) with three substitutions in the VP1 protein-K98E, E145A and L169F-productively infected both mouse cell lines for at least three passages of the virus in murine cells. Moreover, the virus gained the ability to utilize the mSCARB2 protein to infect murine cell lines. These results demonstrate that the three VP1 residues cooperate to effectively interact with the mSCARB2 protein on murine cells and permit the virus to infect murine cells. Gain-of-function studies similar to the present work provide valuable insight into the mutational trajectory required for EV71 to infect new host cells previously non-susceptible to infection.


Asunto(s)
Aminoácidos , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Enterovirus Humano A/química , Enterovirus Humano A/fisiología , Mutación , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de la Cápside/química , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Enterovirus Humano A/genética , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Genética Inversa , Células Vero
11.
PLoS One ; 9(3): e92719, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24671184

RESUMEN

Since its identification in 1969, Enterovirus 71 (EV71) has been causing periodic outbreaks of infection in children worldwide and most prominently in the Asia-Pacific Region. Understanding the pathogenesis of Enterovirus 71 (EV71) is hampered by the virus's inability to infect small animals and replicate in their derived in vitro cultured cells. This manuscript describes the phenotypic and genotypic characteristics of two selected EV71 strains (EV71:TLLm and EV71:TLLmv), which have been adapted to replicate in mouse-derived NIH/3T3 cells, in contrast to the original parental virus which is only able to replicate in primate cell lines. The EV71:TLLm strain exhibited productive infection in all primate and rodent cell lines tested, while EV71:TLLmv exhibited greater preference for mouse cell lines. EV71:TLLmv displayed higher degree of adaptation and temperature adaptability in NIH/3T3 cells than in Vero cells, suggesting much higher fitness in NIH/3T3 cells. In comparison with the parental EV71:BS strain, the adapted strains accumulated multiple adaptive mutations in the genome resulting in amino acid substitutions, most notably in the capsid-encoding region (P1) and viral RNA-dependent RNA polymerase (3D). Two mutations, E167D and L169F, were mapped to the VP1 canyon that binds the SCARB2 receptor on host cells. Another two mutations, S135T and K140I, were located in the VP2 neutralization epitope spanning amino acids 136-150. This is the first report of human EV71 with the ability to productively infect rodent cell lines in vitro.


Asunto(s)
Adaptación Fisiológica , Enterovirus Humano A/genética , Enterovirus Humano A/fisiología , Animales , Antígenos Virales/inmunología , Efecto Citopatogénico Viral , Enterovirus Humano A/crecimiento & desarrollo , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/virología , Genoma Viral , Genotipo , Humanos , Cinética , Ratones , Mutación Missense/genética , Células 3T3 NIH , Fenotipo , Primates , ARN Viral/metabolismo , Temperatura , Transfección , Carga Viral
12.
Emerg Microbes Infect ; 3(2): e15, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26038510

RESUMEN

Saffold virus (SAFV), a newly discovered human cardiovirus of the Picornaviridae family, causes widespread infection among children, as shown by previous seroprevalence studies. To determine the host cell range of SAFV and its cytopathogenicity, eight mammalian cell lines that were available in the laboratory were screened for productive SAFV infection by a laboratory-adapted SAFV of genotype 3. Five of the cell lines (Neuro2A, CHO-K1, NIH/3T3, Vero and HEp-2) were found to be permissible. The time required for SAFV to induce complete lysis as a cytopathic effect (CPE) in these permissibly infected cells and the resultant end point virus titer differed for each cell type. HEp-2 exhibited the shortest time frame to reach full CPE compared to the others. All infected cell lines produced a high virus titer at 72 h post-infection. In addition to causing lytic cell death, SAFV also induced apoptotic cell death in host cells through both extrinsic and intrinsic pathways, although the apoptotic events in HEp-2 cells appeared to have been blocked between the early and late stages. In conclusion, laboratory-adapted SAFV is able to productively infect a number of mammalian cell lines and induce apoptosis in the infected host cells. However, apoptosis in HEp-2 cells is blocked before the end stage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...