Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
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 ; 14(4)2022 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-35458567

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus that caused the coronavirus disease 2019 (COVID-19) pandemic. Though previous studies have suggested that SARS-CoV-2 cellular tropism depends on the host-cell-expressed proteins, whether transcriptional regulation controls SARS-CoV-2 tropism factors in human lung cells remains unclear. In this study, we used computational approaches to identify transcription factors (TFs) regulating SARS-CoV-2 tropism for different types of lung cells. We constructed transcriptional regulatory networks (TRNs) controlling SARS-CoV-2 tropism factors for healthy donors and COVID-19 patients using lung single-cell RNA-sequencing (scRNA-seq) data. Through differential network analysis, we found that the altered regulatory role of TFs in the same cell types of healthy and SARS-CoV-2-infected networks may be partially responsible for differential tropism factor expression. In addition, we identified the TFs with high centralities from each cell type and proposed currently available drugs that target these TFs as potential candidates for the treatment of SARS-CoV-2 infection. Altogether, our work provides valuable cell-type-specific TRN models for understanding the transcriptional regulation and gene expression of SARS-CoV-2 tropism factors.


Asunto(s)
COVID-19 , Redes Reguladoras de Genes , SARS-CoV-2 , Tropismo Viral , Humanos , Pulmón/metabolismo , SARS-CoV-2/genética , Factores de Transcripción/genética , Tropismo Viral/genética
4.
Medicine (Baltimore) ; 101(49): e32024, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36626483

RESUMEN

Human immunodeficiency virus (HIV) infection affects around 37 million people worldwide, and in Kenya, key populations especially female sex workers (FSW), are thought to play a substantial role in the wider, mostly heterosexual HIV-1 transmission structure. Notably, HIV tropism has been found to correlate with HIV-1 transmission and disease progression in HIV-infected patients. In this study, recently infected FSWs from Nairobi, Kenya, were assessed for HIV tropism and the factors related to it. We used a cross-sectional study design to analyze 76 HIV-1 positive plasma samples obtained from FSWs enrolled in sex worker outreach program clinics in Nairobi between November 2020 and April 2021. The effects of clinical, demographic, and viral genetic characteristics were determined using multivariable logistic regression. HIV-1 subtype A1 accounted for 89.5% of all cases, with a prevalence of CXCR4-tropic viruses of 26.3%. WebPSSMR5X4 and Geno2Pheno [G2P:10-15% false positive rate] showed high concordance of 88%. Subjects infected with CXCR4-tropic viruses had statistically significant lower baseline CD4+T-cell counts than those infected with CCR5-tropic viruses (P = .044). Using multivariable logistic regression and adjusting for potential confounders, we found that net charge, the amino acid at position 22 of the V3 loop, and the geographic location of the subject were associated with tropism. A unit increase in V3 loop's net-charge increased the odds of a virus being CXCR4-tropic by 2.4 times (OR = 2.40, 95%CI = 1.35-5.00, P = .007). Second, amino acid threonine at position 22 of V3 loop increased the odds of a strain being X4 by 55.7 times compared to the alanine which occurred in CCR5-tropic strains (OR = 55.7, 95%CI = 4.04-84.1, P < .003). The Kawangware sex worker outreach program clinic was associated with CXCR4-tropic strains (P = .034), but there was there was no evidence of a distinct CXCR4-tropic transmission cluster. In conclusion, this study revealed a high concordance of WebPSSMR5X4 and Geno2Pheno in predicting HIV tropism. The most striking finding was that amino acid position 22 of the V3 loop is linked to tropism in HIV-1 subtype A1. Additional studies with a large dataset are warranted to confirm our findings.


Asunto(s)
Infecciones por VIH , VIH-1 , Trabajadores Sexuales , Tropismo Viral , Femenino , Humanos , Aminoácidos , Estudios Transversales , Infecciones por VIH/epidemiología , VIH-1/genética , Kenia/epidemiología , Receptores CXCR4/metabolismo , Tropismo Viral/genética
5.
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
6.
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
7.
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
9.
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
10.
J Virol ; 95(13): e0217720, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33883222

RESUMEN

Molecular interactions of the variable envelope gp120 subunit of HIV-1 with two cellular receptors are the first step of viral infection, thereby playing pivotal roles in determining viral infectivity and cell tropism. However, the underlying regulatory mechanisms for interactions under gp120 spontaneous variations largely remain unknown. Here, we show an allosteric mechanism in which a single gp120 mutation remotely controls the ternary interactions between gp120 and its receptors for the switch of viral cell tropism. Virological analyses showed that a G310R substitution at the tip of the gp120 V3 loop selectively abolished the viral replication ability in human cells, despite evoking enhancement of viral replication in macaque cells. Molecular dynamics (MD) simulations predicted that the G310R substitution at a site away from the CD4 interaction site selectively impeded the binding ability of gp120 to human CD4. Consistently, virions with the G310R substitution exhibited a reduced binding ability to human lymphocyte cells. Furthermore, the G310R substitution influenced the gp120-CCR5 interaction in a CCR5-type dependent manner as assessed by MD simulations and an infectivity assay using exogenously expressed CCR5s. Interestingly, an I198M mutation in human CCR5 restored the infectivity of the G310R virus in human cells. Finally, MD simulation predicted amino acid interplays that physically connect the V3 loop and gp120 elements for the CD4 and CCR5 interactions. Collectively, these results suggest that the V3 loop tip is a cis-allosteric regulator that remotely controls intra- and intermolecular interactions of HIV-1 gp120 for balancing ternary interactions with CD4 and CCR5. IMPORTANCE Understanding the molecular bases for viral entry into cells will lead to the elucidation of one of the major viral survival strategies, and thus to the development of new effective antiviral measures. As shown recently, HIV-1 is highly mutable and adaptable in growth-restrictive cells, such as those of macaque origin. HIV-1 initiates its infection by sequential interactions of Env-gp120 with two cell surface receptors, CD4 and CCR5. A recent epoch-making structural study has disclosed that CD4-induced conformation of gp120 is stabilized upon binding of CCR5 to the CD4-gp120 complex, whereas the biological significance of this remains totally unknown. Here, from a series of mutations found in our extensive studies, we identified a single-amino acid adaptive mutation at the V3 loop tip of Env-gp120 critical for its interaction with both CD4 and CCR5 in a host cell species-specific way. This remarkable finding could certainly provoke and accelerate studies to precisely clarify the HIV-1 entry mechanism.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/genética , Receptores Virales/metabolismo , Tropismo Viral/genética , Sustitución de Aminoácidos/genética , Animales , Antígenos CD4/metabolismo , Línea Celular , Células HEK293 , VIH-1/patogenicidad , Células HeLa , Humanos , Linfocitos/virología , Macaca fascicularis , Simulación de Dinámica Molecular , Receptores CCR5/metabolismo , Especificidad de la Especie
11.
Methods Mol Biol ; 2244: 19-38, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33555580

RESUMEN

Human cytomegalovirus is routinely isolated by inoculating fibroblast cultures with clinical specimens suspected of harboring HCMV and then monitoring the cultures for cytopathic effects characteristic of this virus. Initially, such clinical isolates are usually strictly cell-associated, but continued propagation in cell culture increases the capacity of an HCMV isolate to release cell-free infectious progeny. Once cell-free infection is possible, genetically homogenous virus strains can be purified by limiting dilution infections. HCMV strains can differ greatly with regard to the titers that can be achieved, the tropism for certain cell types, and the degree to which nonessential genes have been lost during propagation. As there is no ideal HCMV strain for all purposes, the choice of the most appropriate strain depends on the requirements of the particular experiment or project. In this chapter, we provide information that can serve as a basis for deciding which strain may be the most appropriate for a given experiment.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Citomegalovirus/genética , Tropismo Viral/genética , Citomegalovirus/clasificación , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/virología , Fibroblastos/citología , Humanos , Proyectos de Investigación , Tropismo Viral/fisiología , Replicación Viral
12.
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
13.
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
14.
Viruses ; 13(1)2021 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-33477379

RESUMEN

Porcine deltacoronavirus (PDCoV) strain OH-FD22 infects poultry and shares high nucleotide identity with sparrow-origin deltacoronaviruses (SpDCoV) ISU73347 and HKU17 strains. We hypothesized that the spike (S) protein or receptor-binding domain (RBD) from these SpDCoVs would alter the host and tissue tropism of PDCoV. First, an infectious cDNA clone of PDCoV OH-FD22 strain (icPDCoV) was generated and used to construct chimeric icPDCoVs harboring the S protein of HKU17 (icPDCoV-SHKU17) or the RBD of ISU73347 (icPDCoV-RBDISU). To evaluate their pathogenesis, neonatal gnotobiotic pigs were inoculated orally/oronasally with the recombinant viruses or PDCoV OH-FD22. All pigs inoculated with icPDCoV or OH-FD22 developed severe diarrhea and shed viral RNA at moderate-high levels (7.62-10.56 log10 copies/mL) in feces, and low-moderate levels in nasal swabs (4.92-8.48 log10 copies/mL). No pigs in the icPDCoV-SHKU17 and icPDCoV-RBDISU groups showed clinical signs. Interestingly, low-moderate levels (5.07-7.06 log10 copies/mL) of nasal but not fecal viral RNA shedding were detected transiently at 1-4 days post-inoculation in 40% (2/5) of icPDCoV-SHKU17- and 50% (1/2) of icPDCoV-RBDISU-inoculated pigs. These results confirm that PDCoV infected both the upper respiratory and intestinal tracts of pigs. The chimeric viruses displayed an attenuated phenotype with the loss of tropism for the pig intestine. The SpDCoV S protein and RBD reduced viral replication in pigs, suggesting limited potential for cross-species spillover upon initial passage.


Asunto(s)
Infecciones por Coronavirus/patología , Deltacoronavirus/genética , Intestinos/patología , Sistema Respiratorio/patología , Glicoproteína de la Espiga del Coronavirus/genética , Tropismo Viral/genética , Secuencias de Aminoácidos , Animales , Enfermedades de las Aves/virología , Línea Celular , Deltacoronavirus/patogenicidad , Intestinos/virología , Proteínas Recombinantes/genética , Sistema Respiratorio/virología , Gorriones , Porcinos , Enfermedades de los Porcinos/virología , Virulencia/genética
15.
J Pediatric Infect Dis Soc ; 10(1): 1-6, 2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-31981458

RESUMEN

BACKGROUND: Maraviroc, a C-C chemokine receptor 5 (CCR5) antagonist, has been used as an alternative antiretroviral drug in treatment-experienced adults and children infected by CCR5-tropic human immunodeficiency virus type 1 (HIV-1) isolates. Prior to widespread use of this drug, rates of HIV-1 coreceptor tropism and factors associated with coreceptor tropism had to be determined. METHODS: HIV-1-infected individuals aged <20 years with HIV-1 viral loads >1000 RNA copies/mL who were treatment-experienced or treatment-naive were enrolled. HIV-1 coreceptor tropism was determined using a genotypic test in which V3 sequences were analyzed with GENO2PHENO version 2.5 and a false discovery rate of 5%. RESULTS: Fifty-two HIV-1-infected patients were recruited. The median age of participants was 14.9 years (interquartile range [IQR], 8.9-16.8 years). The median CD4 cell count was 396.0 cells/µL (IQR, 72.0-630.3 cells/µL). The median HIV-1 viral load was 43 339 RNA copies/mL (IQR, 8874-197 055 copies/mL). Thirty-nine patients (75%) were treatment-experienced. The most prevalent HIV-1 subtype in this population was CRF01_AE (36 patients, 69.2%). Based on analyses of V3 loop sequences, 5 of 13 treatment-naive patients (38.5%) and 11 of 39 treatment-experienced patients (28.2%) were infected by R5 viruses, while 7 of 13 treatment-naive patients (53.8%) and 19 of 39 treatment-experienced patients (48.7%) were infected by X4 viruses. The only factor associated with the presence of X4 viruses was HIV-1 subtype CRF01_AE. CONCLUSIONS: X4-tropic viruses are associated with the CRF01_AE subtype. Hence, testing of HIV tropism should be performed before treatment with CCR5 inhibitors in children in areas where CRF01_AE predominates.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Tropismo Viral , Adolescente , Recuento de Linfocito CD4 , Niño , Preescolar , Estudios Transversales , Femenino , Técnicas de Genotipaje , Proteína gp120 de Envoltorio del VIH/genética , Infecciones por VIH/epidemiología , VIH-1/genética , Humanos , Masculino , Fragmentos de Péptidos/genética , Tailandia/epidemiología , Carga Viral , Tropismo Viral/genética
16.
Viruses ; 13(1)2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33375604

RESUMEN

Tripartite-motif-containing protein 5 isoform α (TRIM5α) is a cytoplasmic antiretroviral effector upregulated by type I interferons (IFN-I). We previously showed that two points mutations, R332G/R335G, in the retroviral capsid-binding region confer human TRIM5α the capacity to target and strongly restrict HIV-1 upon overexpression of the mutated protein. Here, we used clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-mediated homology-directed repair (HDR) to introduce these two mutations in the endogenous human TRIM5 gene. We found 6 out of 47 isolated cell clones containing at least one HDR-edited allele. One clone (clone 6) had both alleles containing R332G, but only one of the two alleles containing R335G. Upon challenge with an HIV-1 vector, clone 6 was significantly less permissive compared to unmodified cells, whereas the cell clones with monoallelic modifications were only slightly less permissive. Following interferon (IFN)-ß treatment, inhibition of HIV-1 infection in clone 6 was significantly enhanced (~40-fold inhibition). TRIM5α knockdown confirmed that HIV-1 was inhibited by the edited TRIM5 gene products. Quantification of HIV-1 reverse transcription products showed that inhibition occurred through the expected mechanism. In conclusion, we demonstrate the feasibility of potently inhibiting a viral infection through the editing of innate effector genes. Our results also emphasize the importance of biallelic modification in order to reach significant levels of inhibition by TRIM5α.


Asunto(s)
Edición Génica , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/fisiología , Linfocitos T/metabolismo , Linfocitos T/virología , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Tropismo Viral/genética , Factores de Restricción Antivirales , Sistemas CRISPR-Cas , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Patógeno/genética , Humanos , Células Jurkat , ARN Guía de Kinetoplastida , Linfocitos T/inmunología
17.
PLoS Genet ; 16(12): e1009272, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33332358

RESUMEN

The Betacoronaviruses comprise multiple subgenera whose members have been implicated in human disease. As with SARS, MERS and now SARS-CoV-2, the origin and emergence of new variants are often attributed to events of recombination that alter host tropism or disease severity. In most cases, recombination has been detected by searches for excessively similar genomic regions in divergent strains; however, such analyses are complicated by the high mutation rates of RNA viruses, which can produce sequence similarities in distant strains by convergent mutations. By applying a genome-wide approach that examines the source of individual polymorphisms and that can be tested against null models in which recombination is absent and homoplasies can arise only by convergent mutations, we examine the extent and limits of recombination in Betacoronaviruses. We find that recombination accounts for nearly 40% of the polymorphisms circulating in populations and that gene exchange occurs almost exclusively among strains belonging to the same subgenus. Although experimental studies have shown that recombinational exchanges occur at random along the coronaviral genome, in nature, they are vastly overrepresented in regions controlling viral interaction with host cells.


Asunto(s)
Betacoronavirus/clasificación , Betacoronavirus/genética , Recombinación Genética/genética , Glicoproteína de la Espiga del Coronavirus/genética , Intercambio Genético/genética , Genes Virales/genética , Genoma Viral/genética , Especificidad del Huésped/genética , Modelos Genéticos , Polimorfismo Genético , SARS-CoV-2/clasificación , SARS-CoV-2/genética , Tropismo Viral/genética
18.
Virol J ; 17(1): 169, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33143744

RESUMEN

BACKGROUND: In Tianjin, China, there is a relatively high prevalence of HIV in men who have sex with men (MSM). The number of HIV cases in Tianjin is also increasing. We investigated the HIV molecular transmission network, genetic tropisms, and drug resistance mutations in Tianjin. METHODS: Blood samples were collected from 510 newly diagnosed antiretroviral therapy (ART)-naïve HIV-1-infected subjects among MSM in Tianjin. Partial pol and env genes were sequenced and used for phylogenetic, genetic tropism, and genotypic drug resistance analyses. Molecular clusters were identified with 1.5% genetic distance and 90% bootstrap support. RESULTS: Among the 436 HIV-1 pol sequences obtained from the study participants, various genotypes were identified, including CRF01_AE (56.9%), CRF07_BC (27.8%), B (7.3%), CRF55_01B (4.1%), unique recombinant forms (URFs) (3.7%), and CRF59_01B (0.2%). A higher prevalence of X4 viruses was observed in individuals infected with CRF55_01B (56.3%) and CRF01_AE (46.2%) than with other subtypes. Of all 110 sequences in the 36 clusters, 62 (56.4%) were observed in 23 CRF01_AE clusters and 18 (16.4%) in four CRF07_BC clusters. Eight sequences clustered with at least one other shared the same drug resistance mutation (DRM). In different cluster sizes, the distributions of individuals by age, presence of sexually transmitted disease, and presence of DRMs, were significantly different. CONCLUSION: We revealed the characteristics of HIV molecular transmission, tropism, and DRMs of ART-naïve HIV-infected individuals among the MSM population in Tianjin. Identifying infected persons at risk of transmission is necessary for proposing counseling and treating these patients to reduce the risk of HIV transmission.


Asunto(s)
Farmacorresistencia Viral/genética , Genotipo , Infecciones por VIH/transmisión , VIH-1/efectos de los fármacos , VIH-1/genética , Homosexualidad Masculina/estadística & datos numéricos , Adolescente , Adulto , Anciano , Genes env/genética , Infecciones por VIH/sangre , Infecciones por VIH/virología , VIH-1/clasificación , Humanos , Masculino , Persona de Mediana Edad , Mutación , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Tropismo Viral/genética , Adulto Joven
19.
Commun Biol ; 3(1): 559, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033372

RESUMEN

Hyper-virulent Streptococcus pneumoniae serotype 1 strains are endemic in Sub-Saharan Africa and frequently cause lethal meningitis outbreaks. It remains unknown whether genetic variation in serotype 1 strains modulates tropism into cerebrospinal fluid to cause central nervous system (CNS) infections, particularly meningitis. Here, we address this question through a large-scale linear mixed model genome-wide association study of 909 African pneumococcal serotype 1 isolates collected from CNS and non-CNS human samples. By controlling for host age, geography, and strain population structure, we identify genome-wide statistically significant genotype-phenotype associations in surface-exposed choline-binding (P = 5.00 × 10-08) and helicase proteins (P = 1.32 × 10-06) important for invasion, immune evasion and pneumococcal tropism to CNS. The small effect sizes and negligible heritability indicated that causation of CNS infection requires multiple genetic and other factors reflecting a complex and polygenic aetiology. Our findings suggest that certain pathogen genetic variation modulate pneumococcal survival and tropism to CNS tissue, and therefore, virulence for meningitis.


Asunto(s)
Variación Genética/genética , Meningitis Neumocócica/microbiología , Streptococcus pneumoniae/patogenicidad , Tropismo Viral/genética , Adolescente , Sistema Nervioso Central/microbiología , Niño , Preescolar , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/fisiología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...