Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
J Virol ; 98(2): e0150423, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38289119

RESUMEN

Coxsackievirus B3 (CVB3) is known to cause acute myocarditis and pancreatitis in humans. We investigated the microRNAs (miRNAs) that can potentially govern the viral life cycle by binding to the untranslated regions (UTRs) of CVB3 RNA. MicroRNA-22-3p was short-listed, as its potential binding site overlapped with the region crucial for recruiting internal ribosome entry site trans-acting factors (ITAFs) and ribosomes. We demonstrate that miR-22-3p binds CVB3 5' UTR, hinders recruitment of key ITAFs on viral mRNA, disrupts the spatial structure required for ribosome recruitment, and ultimately blocks translation. Likewise, cells lacking miR-22-3p exhibited heightened CVB3 infection compared to wild type, confirming its role in controlling infection. Interestingly, miR-22-3p level was found to be increased at 4 hours post-infection, potentially due to the accumulation of viral 2A protease in the early phase of infection. 2Apro enhances the miR-22-3p level to dislodge the ITAFs from the SD-like sequence, rendering the viral RNA accessible for binding of replication factors to switch to replication. Furthermore, one of the cellular targets of miR-22-3p, protocadherin-1 (PCDH1), was significantly downregulated during CVB3 infection. Partial silencing of PCDH1 reduced viral replication, demonstrating its proviral role. Interestingly, upon CVB3 infection in mice, miR-22-3p level was found to be downregulated only in the small intestine, the primary target organ, indicating its possible role in influencing tissue tropism. It appears miR-22-3p plays a dual role during infection by binding viral RNA to aid its life cycle as a viral strategy and by targeting a proviral protein to restrict viral replication as a host response.IMPORTANCECVB3 infection is associated with the development of end-stage heart diseases. Lack of effective anti-viral treatments and vaccines for CVB3 necessitates comprehensive understanding of the molecular players during CVB3 infection. miRNAs have emerged as promising targets for anti-viral strategies. Here, we demonstrate that miR-22-3p binds to 5' UTR and inhibits viral RNA translation at the later stage of infection to promote viral RNA replication. Conversely, as host response, it targets PCDH1, a proviral factor, to discourage viral propagation. miR-22-3p also influences CVB3 tissue tropism. Deciphering the multifaced role of miR-22-3p during CVB3 infection unravels the necessary molecular insights, which can be exploited for novel intervening strategies to curb infection and restrict viral pathogenesis.


Asunto(s)
Regiones no Traducidas 5' , Infecciones por Coxsackievirus , Enterovirus Humano B , Interacciones Microbiota-Huesped , MicroARNs , Biosíntesis de Proteínas , ARN Viral , Animales , Humanos , Ratones , Regiones no Traducidas 5'/genética , Antivirales/metabolismo , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/virología , Enterovirus Humano B/genética , Enterovirus Humano B/patogenicidad , Enterovirus Humano B/fisiología , Células HeLa , Intestino Delgado/metabolismo , Intestino Delgado/virología , MicroARNs/genética , MicroARNs/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Tropismo Viral/genética , Replicación Viral/genética , Cisteína Endopeptidasas/metabolismo , Protocadherinas/deficiencia , Protocadherinas/genética , Miocarditis , Interacciones Microbiota-Huesped/genética
2.
Nat Biotechnol ; 41(9): 1272-1286, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36702899

RESUMEN

A barrier to advancing engineered adeno-associated viral vectors (AAVs) for precision access to cell subtypes is a lack of high-throughput, high-resolution assays to characterize in vivo transduction profiles. In this study, we developed an ultrasensitive, sequential fluorescence in situ hybridization (USeqFISH) method for spatial transcriptomic profiling of endogenous and viral RNA with a short barcode in intact tissue volumes by integrating hydrogel-based tissue clearing, enhanced signal amplification and multiplexing using sequential labeling. Using USeqFISH, we investigated the transduction and cell subtype tropisms across mouse brain regions of six systemic AAVs, including AAV-PHP.AX, a new variant that transduces robustly and efficiently across neurons and astrocytes. Here we reveal distinct cell subtype biases of each AAV variant, including a bias of AAV-PHP.N toward excitatory neurons. USeqFISH also enables profiling of pooled regulatory cargos, as we show for a 13-variant pool of microRNA target sites in AAV genomes. Lastly, we demonstrate potential applications of USeqFISH for in situ AAV profiling and multimodal single-cell analysis in non-human primates.


Asunto(s)
Técnicas de Transferencia de Gen , Transcriptoma , Ratones , Animales , Transducción Genética , Hibridación Fluorescente in Situ , Transcriptoma/genética , Vectores Genéticos/genética , Tropismo/genética , Dependovirus/genética , Tropismo Viral/genética
3.
Viruses ; 15(1)2022 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-36680147

RESUMEN

HIV/AIDS mortality has been decreasing over the last decade. While promising, this decrease correlated directly with increased use of antiretroviral drugs. As a natural consequence of its high mutation rate, treatments provide selection pressure that promotes the natural selection of escape mutants. Individuals may acquire drug-naive strains, or those that have already mutated due to treatment. Even within a host, mutation affects HIV tropism, where initial infection begins with R5-tropic virus, but the clinical transition to AIDS correlates with mutations that lead to an X4-tropic switch. Furthermore, the high mutation rate of HIV has spelled failure for all attempts at an effective vaccine. Pre-exposure drugs are currently the most effective drug-based preventatives, but their effectiveness is also threatened by viral mutation. From attachment and entry to assembly and release, the steps in the replication cycle are also discussed to describe the drug mechanisms and mutations that arise due to those drugs. Revealing the patterns of HIV-1 mutations, their effects, and the coordinated attempt to understand and control them will lead to effective use of current preventative measures and treatment options, as well as the development of new ones.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH , Humanos , Infecciones por VIH/tratamiento farmacológico , Mutación , Tropismo Viral/genética , Replicación Viral , VIH/efectos de los fármacos , VIH/genética , Fármacos Anti-VIH/uso terapéutico
4.
Biol Open ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34494647

RESUMEN

Ex vivo, gene therapy is a powerful approach holding great promises for the treatment of both genetic and acquired diseases. Adeno-associated virus (AAV) vectors are a safe and efficient delivery system for modification of mesenchymal stem cells (MSC) that could maximize their therapeutic benefits. Assessment of MSC viability and functional activity after infection with new AAV serotypes is necessary, due to AAV tropism to specific cell types. We infected human and rat adipose-tissue MSC with hybrid AAV-DJ serotype vectors carrying GFP and SCF genes. GFP expression from AAV-DJ was about 1.5-fold superior to that observed with AAV-2 and lasted for at least 21 days as was evaluated by flow cytometry and fluorescence microscopy. AAV-DJ proves to be suitable for the infection of rat and human MSC with a similar efficiency. Infected MSC were still viable but showed a 25-30% growth-rate slowdown. Moreover, we found an increase of SERPINB2 mRNA expression in human MSC while expression of other oxidative stress markers and extracellular matrix proteins was not affected. These results suggest that there is a differential cellular response in MSC infected with AAV viral vectors, which should be taken into account as it can affect the expected outcome for the therapeutic application.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/sangre , Células Madre Mesenquimatosas/virología , Proteínas Virales/sangre , Animales , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratas , Serogrupo , Factor de Células Madre/metabolismo , Tropismo Viral/genética
5.
IUBMB Life ; 73(8): 1005-1015, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34118117

RESUMEN

The kidney is one of the main targets attacked by viruses in patients with a coronavirus infection. Until now, SARS-CoV-2 has been identified as the seventh member of the coronavirus family capable of infecting humans. In the past two decades, humankind has experienced outbreaks triggered by two other extremely infective members of the coronavirus family; the MERS-CoV and the SARS-CoV. According to several investigations, SARS-CoV causes proteinuria and renal impairment or failure. The SARS-CoV was identified in the distal convoluted tubules of the kidney of infected patients. Also, renal dysfunction was observed in numerous cases of MERS-CoV infection. And recently, during the 2019-nCoV pandemic, it was found that the novel coronavirus not only induces acute respiratory distress syndrome (ARDS) but also can induce damages in various organs including the liver, heart, and kidney. The kidney tissue and its cells are targeted massively by the coronaviruses due to the abundant presence of ACE2 and Dpp4 receptors on kidney cells. These receptors are characterized as the main route of coronavirus entry to the victim cells. Renal failure due to massive viral invasion can lead to undesirable complications and enhanced mortality rate, thus more attention should be paid to the pathology of coronaviruses in the kidney. Here, we have provided the most recent knowledge on the coronaviruses (SARS, MERS, and COVID19) pathology and the mechanisms of their impact on the kidney tissue and functions.


Asunto(s)
COVID-19/mortalidad , Infecciones por Coronavirus/mortalidad , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , SARS-CoV-2/patogenicidad , Síndrome Respiratorio Agudo Grave/mortalidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Tropismo Viral/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/genética , COVID-19/patología , COVID-19/virología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Regulación de la Expresión Génica , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Unión Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/patología , Síndrome Respiratorio Agudo Grave/virología , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Análisis de Supervivencia
7.
Viruses ; 13(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916225

RESUMEN

Measles virus (MV) can cause severe acute diseases as well as long-lasting clinical deteriorations due to viral-induced immunosuppression and neuronal manifestation. How the virus enters the brain and manages to persist in neuronal tissue is not fully understood. Various mutations in the viral genes were found in MV strains isolated from patient brains. In this study, reverse genetics was used to introduce mutations in the fusion, matrix and polymerase genes of MV. The generated virus clones were characterized in cell culture and used to infect rat brain slice cultures. A mutation in the carboxy-terminal domain of the matrix protein (R293Q) promoted the production of progeny virions. This effect was observed in Vero cells irrespective of the expression of the signaling lymphocyte activation molecule (SLAM). Furthermore, a mutation in the fusion protein (I225M) induced syncytia formation on Vero cells in the absence of SLAM and promoted viral spread throughout the rat brain slices. In this study, a solid ex vivo model was established to elucidate the MV mutations contributing to neural manifestation.


Asunto(s)
Encéfalo/virología , Virus del Sarampión/genética , Mutación , Neuronas/virología , Proteínas Virales/genética , Tropismo Viral/genética , Animales , Chlorocebus aethiops , Células HEK293 , Humanos , Técnicas In Vitro , Sarampión/virología , Virus del Sarampión/patogenicidad , Virus del Sarampión/fisiología , Ratas Endogámicas Lew , Genética Inversa , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Células Vero , Proteínas Virales de Fusión/genética
8.
Methods Mol Biol ; 2244: 51-81, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33555582

RESUMEN

The extensive tropism of human cytomegalovirus (HCMV) results in the productive infection of multiple cell types within the human host. However, infection of other cell types, such as undifferentiated cells of the myeloid lineage, give rise to nonpermissive infections. This aspect has been used experimentally to model latent infection, which is known to be established in the pluripotent CD34+ hematopoietic progenitor cell population resident in the bone marrow in vivo. The absence of a tractable animal model for studies of HCMV has resulted in a number of laboratories employing experimental infection of cells in vitro to simulate both HCMV lytic and latent infection. Herein, we will focus on the techniques used in our laboratory for the isolation and use of primary cells to study aspects of HCMV latency, reactivation, and lytic infection.


Asunto(s)
Citomegalovirus/metabolismo , Cultivo Primario de Células/métodos , Antígenos CD34/metabolismo , Diferenciación Celular , Infecciones por Citomegalovirus/virología , Células Madre Hematopoyéticas/metabolismo , Monocitos/metabolismo , Transducción de Señal , Tropismo Viral/genética , Tropismo Viral/fisiología , Activación Viral , Latencia del Virus
9.
Methods Mol Biol ; 2244: 83-101, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33555583

RESUMEN

Of the many research challenges posed by the study of human cytomegalovirus (HCMV) latency, one of the most notable is the requirement for the use of primary hematopoietic cell culture. Culturing hematopoietic progenitor subpopulations requires that consideration be given to maintaining their physiological relevance. We describe a long-standing primary CD34+ hematopoietic progenitor cell (HPC) system as an in vitro model to study HCMV latent infection. Key aspects of the model include infection of primary human CD34+ HPCs prior to ex vivo expansion, a long-term culture with a stromal cell support designed to maintain the ability of stem cells to support hematopoietic reconstitution, and an assay to quantify infectious centers produced prior to and following a reactivation stimulus. Importantly, this system has been used to identify a number of viral determinants of latency or reactivation and findings have been recapitulated in vivo using a humanized mouse model for HCMV latency. Therefore, this system offers a powerful approach to defining virus-host interactions and mechanisms important for HCMV latency and reactivation.


Asunto(s)
Citomegalovirus/metabolismo , Cultivo Primario de Células/métodos , Latencia del Virus/fisiología , Antígenos CD34/metabolismo , Diferenciación Celular , Infecciones por Citomegalovirus/virología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Transducción de Señal , Proteínas Virales , Tropismo Viral/genética , Tropismo Viral/fisiología , Activación Viral/genética , Activación Viral/fisiología
10.
Viruses ; 12(10)2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053912

RESUMEN

Polyomaviruses are ubiquitous human pathogens that cause lifelong, asymptomatic infections in healthy individuals. Although these viruses are restrained by an intact immune system, immunocompromised individuals are at risk for developing severe diseases driven by resurgent viral replication. In particular, loss of immune control over JC polyomavirus can lead to the development of the demyelinating brain disease progressive multifocal leukoencephalopathy (PML). Viral isolates from PML patients frequently carry point mutations in the major capsid protein, VP1, which mediates virion binding to cellular glycan receptors. Because polyomaviruses are non-enveloped, VP1 is also the target of the host's neutralizing antibody response. Thus, VP1 mutations could affect tropism and/or recognition by polyomavirus-specific antibodies. How these mutations predispose susceptible individuals to PML and other JCPyV-associated CNS diseases remains to be fully elucidated. Here, we review the current understanding of polyomavirus capsid mutations and their effects on viral tropism, immune evasion, and virulence.


Asunto(s)
Proteínas de la Cápside/genética , Evasión Inmune/genética , Virus JC/genética , Virus JC/inmunología , Leucoencefalopatía Multifocal Progresiva/inmunología , Animales , Cápside/inmunología , Proteínas de la Cápside/inmunología , Especificidad del Huésped/genética , Humanos , Evasión Inmune/inmunología , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/virología , Ratones , Mutación , Tropismo Viral/genética , Replicación Viral/genética
11.
Hum Gene Ther ; 31(23-24): 1288-1299, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32948113

RESUMEN

Viral-mediated gene augmentation offers tremendous promise for the treatment of inherited retinal diseases. The development of effective gene therapy requires an understanding of the vector's tissue-specific behavior, which may vary depending on serotype, route of delivery, or target species. Using an ex vivo organotypic explant system, we previously demonstrated that retinal tropism and transduction of adeno-associated virus type 2 (AAV2) vary significantly depending on serotype in human eyes. However, the ex vivo system has limited ability to assess route of ocular delivery, and relatively little literature exists on tropic differences between serotypes and routes of delivery in vivo. In this study, we demonstrate that retinal tropism and transduction efficiency of five different AAV2 serotypes (AAV2/1, AAV2/2, AAV2/6, AAV2/8, and AAV2/9) expressing enhanced green fluorescent protein driven by a cytomegalovirus promoter vary greatly depending on serotype and route of delivery (intravitreal, subretinal, or suprachoroidal) in rats. With subretinal delivery, all serotypes successfully transduced the retinal pigmented epithelium and outer nuclear layer (ONL), with AAV2/1 displaying the highest transduction efficiency and AAV2/2 and AAV2/6 showing lower ONL transduction. There was minimal transduction of the inner retina through subretinal delivery for any serotype. Tropism by suprachoroidal delivery mirrored that of subretinal delivery for all AAV serotypes but resulted in a wider distribution and greater ONL transduction. With intravitreal delivery, retinal transduction was seen primarily in the inner retina (retinal nerve fiber, ganglion cell, and inner nuclear layers) for AAV2/1 and AAV2/6, with AAV2/6 showing the highest transduction. When compared with data from human explant models, there are substantial differences in tropism and transduction that are important to consider when using rats as preclinical models for the development of ocular gene therapies for humans.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Enfermedades de la Retina/terapia , Pigmentos Retinianos/genética , Animales , Vías de Administración de Medicamentos , Epitelio/metabolismo , Epitelio/patología , Humanos , Inyecciones Intravítreas , Ratas , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Serogrupo , Líquido Subretiniano , Neuronas del Núcleo Supraquiasmático/metabolismo , Neuronas del Núcleo Supraquiasmático/patología , Tropismo Viral/genética
12.
Viruses ; 12(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32604765

RESUMEN

Geminiviruses and their diseases are a considerable economic threat to a vast number of crops worldwide. Investigating how and where these viruses replicate and accumulate in their hosts may lead to novel molecular resistance strategies. In this study, we used the Rep-inducible In Plant Activation (INPACT) expression platform, based on the genome of tobacco yellow dwarf virus (TYDV), to determine where this model mastrevirus replicates in its host tobacco. By developing an infectious clone of TYDV and optimizing its delivery by agroinfiltration, we first established an efficient artificial infection process. When delivered into transgenic tobacco plants containing a TYDV-based INPACT cassette encoding the ß-glucuronidase (GUS) reporter, we showed the virus activates GUS expression. Histology revealed that reporter gene expression was limited to phloem-associated cell types suggesting TYDV replication has a restricted tissue tropism.


Asunto(s)
Geminiviridae/crecimiento & desarrollo , Regulación de la Expresión Génica de las Plantas/genética , Nicotiana/virología , Tropismo Viral/genética , Replicación Viral/fisiología , Geminiviridae/genética , Genoma Viral/genética , Glucuronidasa/genética , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Enfermedades de las Plantas/virología , Plantas Modificadas Genéticamente/virología , Replicación Viral/genética
13.
Annu Rev Virol ; 7(1): 289-307, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32603631

RESUMEN

Viral infection underlies a significant share of the global cancer burden. Merkel cell polyomavirus (MCPyV) is the newest member of the human oncogenic virus family. Its discovery over a decade ago marked the beginning of an exciting era in human tumor virology. Since then, significant evidence has emerged to support the etiologic role of MCPyV in Merkel cell carcinoma (MCC), an extremely lethal form of skin cancer. MCPyV infection is widespread in the general population. MCC diagnoses have tripled over the past 20 years, but effective treatments are currently lacking. In this review, we highlight recent discoveries that have shaped our understanding of MCPyV oncogenic mechanism and host cellular tropism, as well as the molecular events occurring in the viral infectious life cycle. These insights will guide future efforts in developing novel virus-targeted therapeutic strategies for treating the devastating human cancers associated with this new tumorigenic virus.


Asunto(s)
Carcinogénesis , Poliomavirus de Células de Merkel/genética , Tropismo Viral/genética , Replicación Viral/genética , Carcinoma de Células de Merkel/patología , Carcinoma de Células de Merkel/virología , Genes Virales , Humanos , Poliomavirus de Células de Merkel/metabolismo , Oncogenes , Infecciones por Polyomavirus/complicaciones , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/virología
14.
Viruses ; 12(5)2020 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-32370153

RESUMEN

The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a lethal zoonotic pathogen circulating in the Arabian Peninsula since 2012. There is no vaccine for MERS and anti-viral treatment is generally not applicable. We investigated the evolution of the MERS-CoV spike gene sequences and changes in viral loads over time from patients in Saudi Arabia from 2105-2017. All the MERS-CoV strains belonged to lineage 5, and showed high sequence homology (99.9%) to 2017 strains. Recombination analysis showed a potential recombination event in study strains from patients in Saudi Arabia. The spike gene showed eight amino acid substitutions, especially between the A1 and B5 lineage, and contained positively selected codon 1020. We also determined that the viral loads were significantly (p < 0.001) higher in fatal cases, and virus shedding was prolonged in some fatal cases beyond 21 days. The viral concentration peaked during the first week of illness, and the lower respiratory specimens had higher levels of MERS-CoV RNA. The presence of the diversifying selection and the topologies with the structural mapping of residues under purifying selection suggested that codon 1020 might have a role in the evolution of spike gene during the divergence of different lineages. This study will im-prove our understanding of the evolution of MERS-CoV, and also highlights the need for enhanced surveillance in humans and dromedaries. The presence of amino acid changes at the N-terminal domain and structural mapping of residues under positive selection at heptad repeat 1 provides better insight into the adaptive evolution of the spike gene and might have a potential role in virus-host tropism and pathogenesis.


Asunto(s)
Sustitución de Aminoácidos/genética , Infecciones por Coronavirus/patología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Glicoproteína de la Espiga del Coronavirus/genética , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Camelus/virología , Dipeptidil Peptidasa 4/metabolismo , Evolución Molecular , Femenino , Genoma Viral/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Dominios Proteicos/genética , ARN Viral/genética , Receptores Virales/genética , Recombinación Genética/genética , Arabia Saudita , Análisis de Secuencia de ARN , Homología de Secuencia , Carga Viral , Tropismo Viral/genética
15.
PLoS Pathog ; 15(10): e1008057, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31671153

RESUMEN

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


Asunto(s)
Infecciones por Astroviridae/inmunología , Infecciones por Astroviridae/veterinaria , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Mamastrovirus/fisiología , Tropismo Viral/genética , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Enterocitos/virología , Gastroenteritis/virología , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/virología , Intestino Delgado/citología , Intestino Delgado/virología , Mamastrovirus/genética , Mamastrovirus/inmunología , Células Vero , Tropismo Viral/inmunología
16.
FEBS Lett ; 593(24): 3583-3608, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31769017

RESUMEN

Human adenovirus commonly causes infections of respiratory, gastrointestinal, genitourinary, and ocular surface mucosae. Although most adenovirus eye infections are mild and self-limited, specific viruses within human adenovirus species D are associated with epidemic keratoconjunctivitis (EKC), a severe and highly contagious ocular surface infection, which can lead to chronic and/or recurrent, visually disabling keratitis. In this review, we discuss the links between adenovirus ontogeny, genomics, immune responses, and corneal pathogenesis, for those viruses that cause EKC.


Asunto(s)
Adenovirus Humanos/patogenicidad , Evolución Biológica , Proteínas del Ojo/genética , Interacciones Huésped-Patógeno/genética , Queratitis/genética , Queratoconjuntivitis/genética , Proteínas Virales/genética , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Conjuntiva/inmunología , Conjuntiva/metabolismo , Conjuntiva/patología , Conjuntiva/virología , Córnea/inmunología , Córnea/metabolismo , Córnea/patología , Córnea/virología , Modelos Animales de Enfermedad , Proteínas del Ojo/inmunología , Regulación de la Expresión Génica , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Queratitis/inmunología , Queratitis/patología , Queratitis/virología , Queratoconjuntivitis/inmunología , Queratoconjuntivitis/patología , Queratoconjuntivitis/virología , Filogenia , Proteínas Virales/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología
17.
Virus Res ; 274: 197771, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31577935

RESUMEN

We compared the phenotypes of three mutant AAV2 viruses containing mutations in arginine amino acids (R585, R588 and R484) previously shown to be involved in AAV2 heparan sulfate binding. The transduction efficiencies of wild type and mutant viruses were determined in the eye, the brain and peripheral organs following subretinal, striatal and intravenous injection, respectively, in mice and rats. We found that each of the three mutants (the single mutant R585A; the double mutant R585, 588A; and the triple mutant R585, 588, 484A) had a unique phenotype compared to wt and each other. R585A was completely defective for transducing peripheral organs via intravenous injection, suggesting that R585A may be useful for targeting peripheral organs by substitution of peptide ligands in the capsid surface. In the brain, all three mutants displayed widespread transduction, with the double mutant R585, 588A displaying the greatest spread and the greatest number of transduced neurons. The double mutant was also extremely efficient for retrograde transport, while the triple mutant was almost completely defective for retrograde transport. This suggested that R484 may be directly involved in interaction with the transport machinery. Finally, the double mutant also displayed improved transduction of the eye compared to wild type and the other mutants.


Asunto(s)
Proteínas de la Cápside/genética , Cápside/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Parvovirinae/fisiología , Animales , Transporte Axonal/genética , Proteínas de la Cápside/metabolismo , Dependovirus , Femenino , Masculino , Ratones , Mutación , Parvovirinae/genética , Parvovirinae/metabolismo , Fenotipo , Unión Proteica , Ratas , Tropismo Viral/genética
18.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814288

RESUMEN

Ferrets represent an invaluable animal model to study influenza virus pathogenesis and transmission. To further characterize this model, we developed a differentiated primary ferret nasal epithelial cell (FNEC) culture model for investigation of influenza A virus infection and virus-host interactions. This well-differentiated culture consists of various cell types, a mucociliary clearance system, and tight junctions, representing the nasal ciliated pseudostratified respiratory epithelium. Both α2,6-linked and α2,3-linked sialic acid (SA) receptors, which preferentially bind the hemagglutinin (HA) of human and avian influenza viruses, respectively, were detected on the apical surface of the culture with different cellular tropisms. In accordance with the distribution of SA receptors, we observed that a pre-2009 seasonal A(H1N1) virus infected both ciliated and nonciliated cells, whereas a highly pathogenic avian influenza (HPAI) A(H5N1) virus primarily infected nonciliated cells. Transmission electron microscopy revealed that virions were released from or associated with the apical membranes of ciliated, nonciliated, and mucin-secretory goblet cells. Upon infection, the HPAI A(H5N1) virus replicated to titers higher than those of the human A(H1N1) virus at 37°C; however, replication of the A(H5N1) virus was significantly attenuated at 33°C. Furthermore, we found that infection with the A(H5N1) virus induced higher expression levels of immune mediator genes and resulted in more cell damage/loss than with the human A(H1N1) virus. This primary differentiated FNEC culture model, recapitulating the structure of the nasal epithelium, provides a useful model to bridge in vivo and in vitro studies of cellular tropism, infectivity, and pathogenesis of influenza viruses during the initial stages of infection.IMPORTANCE Although ferrets serve as an important model of influenza virus infection, much remains unknown about virus-host interactions in this species at the cellular level. The development of differentiated primary cultures of ferret nasal epithelial cells is an important step toward understanding cellular tropism and the mechanisms of influenza virus infection and replication in the airway milieu of this model. Using lectin staining and microscopy techniques, we characterized the sialic acid receptor distribution and the cellular composition of the culture model. We then evaluated the replication of and immune response to human and avian influenza viruses at relevant physiological temperatures. Our findings offer significant insight into this first line of defense against influenza virus infection and provide a model for the evaluation of emerging influenza viruses in a well-controlled in vitro environmental setting.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Tropismo Viral/genética , Animales , Bronquios/virología , Técnicas de Cultivo de Célula/métodos , Cilios/virología , Modelos Animales de Enfermedad , Células Epiteliales/virología , Hurones/virología , Células Caliciformes/metabolismo , Células Caliciformes/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Gripe Humana/virología , Mucosa Nasal/metabolismo , Mucosa Nasal/virología , Cultivo Primario de Células , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Mucosa Respiratoria/virología , Tráquea/virología , Virosis/genética
19.
Elife ; 72018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30520725

RESUMEN

Interferon (IFN) inhibits HIV replication by inducing antiviral effectors. To comprehensively identify IFN-induced HIV restriction factors, we assembled a CRISPR sgRNA library of Interferon Stimulated Genes (ISGs) into a modified lentiviral vector that allows for packaging of sgRNA-encoding genomes in trans into budding HIV-1 particles. We observed that knockout of Zinc Antiviral Protein (ZAP) improved the performance of the screen due to ZAP-mediated inhibition of the vector. A small panel of IFN-induced HIV restriction factors, including MxB, IFITM1, Tetherin/BST2 and TRIM5alpha together explain the inhibitory effects of IFN on the CXCR4-tropic HIV-1 strain, HIV-1LAI, in THP-1 cells. A second screen with a CCR5-tropic primary strain, HIV-1Q23.BG505, described an overlapping, but non-identical, panel of restriction factors. Further, this screen also identifies HIV dependency factors. The ability of IFN-induced restriction factors to inhibit HIV strains to replicate in human cells suggests that these human restriction factors are incompletely antagonized. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Asunto(s)
Células Epiteliales/inmunología , Edición Génica/métodos , VIH-1/genética , Interacciones Huésped-Patógeno , Proteínas Nucleares/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Factores de Restricción Antivirales , Sistemas CRISPR-Cas , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/inmunología , Células HEK293 , VIH-1/efectos de los fármacos , VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Humanos , Interferón-alfa/farmacología , Lentivirus/genética , Lentivirus/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/inmunología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/inmunología , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/inmunología , Proteínas de Unión al ARN , Receptores CCR5/genética , Receptores CCR5/inmunología , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Proteínas Represoras , Transducción de Señal , Células THP-1 , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Tropismo Viral/genética , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
20.
J Virol ; 92(18)2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29997207

RESUMEN

UL148 is a viral endoplasmic reticulum (ER)-resident glycoprotein that contributes to human cytomegalovirus (HCMV) cell tropism. The influence of UL148 on tropism correlates with its potential to promote the expression of glycoprotein O (gO), a viral envelope glycoprotein that participates in a heterotrimeric complex with glycoproteins H and L that is required for infectivity. In an effort to gain insight into the mechanism, we used mass spectrometry to identify proteins that coimmunoprecipitate from infected cells with UL148. This approach led us to identify an interaction between UL148 and SEL1L, a factor that plays key roles in ER-associated degradation (ERAD). In pulse-chase experiments, gO was less stable in cells infected with UL148-null mutant HCMV than during wild-type infection, suggesting a potential functional relevance for the interaction with SEL1L. To investigate whether UL148 regulates gO abundance by influencing ERAD, small interfering RNA (siRNA) silencing of either SEL1L or its partner, Hrd1, was carried out in the context of infection. Knockdown of these ERAD factors strongly enhanced levels of gO but not other viral glycoproteins, and the effect was amplified in the presence of UL148. Furthermore, pharmacological inhibition of ERAD showed similar results. Silencing of SEL1L during infection also stabilized an interaction of gO with the ER lectin OS-9, which likewise suggests that gO is an ERAD substrate. Taken together, our results identify an intriguing interaction of UL148 with the ERAD machinery and demonstrate that gO behaves as a constitutive ERAD substrate during infection. These findings have implications for understanding the regulation of HCMV cell tropism.IMPORTANCE Viral glycoproteins in large part determine the cell types that an enveloped virus can infect and hence play crucial roles in transmission and pathogenesis. The glycoprotein H/L heterodimer (gH/gL) is part of the conserved membrane fusion machinery that all herpesviruses use to enter cells. In human cytomegalovirus (HCMV), gH/gL participates in alternative complexes in virions, one of which is a trimer of gH/gL with glycoprotein O (gO). Here, we show that gO is constitutively degraded during infection by the endoplasmic reticulum-associated degradation (ERAD) pathway and that UL148, a viral factor that regulates HCMV cell tropism, interacts with the ERAD machinery and slows gO decay. Since gO is required for cell-free virus to enter new host cells but dispensable for cell-associated spread that resists antibody neutralization, our findings imply that the posttranslational instability of a viral glycoprotein provides a basis for viral mechanisms to modulate tropism and spread.


Asunto(s)
Citomegalovirus/genética , Retículo Endoplásmico/virología , Glicoproteínas de Membrana/genética , Proteínas/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética , Tropismo Viral/genética , Células Cultivadas , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Retículo Endoplásmico/fisiología , Células Epiteliales/virología , Fibroblastos/virología , Regulación de la Expresión Génica , Humanos , Mutación con Pérdida de Función , Espectrometría de Masas , Glicoproteínas de Membrana/metabolismo , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Tropismo Viral/fisiología , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA