Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 396
Filtrar
2.
Int J Mol Sci ; 23(3)2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35163212

RESUMEN

Cell death by apoptosis is a major cellular response in the control of tissue homeostasis and as a defense mechanism in the case of cellular aggression such as an infection. Cell self-destruction is part of antiviral responses, aimed at limiting the spread of a virus. Although it may contribute to the deleterious effects in infectious pathology, apoptosis remains a key mechanism for viral clearance and the resolution of infection. The control mechanisms of cell death processes by viruses have been extensively studied. Apoptosis can be triggered by different viral determinants through different pathways as a result of virally induced cell stresses and innate immune responses. Zika virus (ZIKV) induces Zika disease in humans, which has caused severe neurological forms, birth defects, and microcephaly in newborns during the last epidemics. ZIKV also surprised by revealing an ability to persist in the genital tract and in semen, thus being sexually transmitted. Mechanisms of diverting antiviral responses such as the interferon response, the role of cytopathic effects and apoptosis in the etiology of the disease have been widely studied and debated. In this review, we examined the interplay between ZIKV infection of different cell types and apoptosis and how the virus deals with this cellular response. We illustrate a duality in the effects of ZIKV-controlled apoptosis, depending on whether it occurs too early or too late, respectively, in neuropathogenesis, or in long-term viral persistence. We further discuss a prospective role for apoptosis in ZIKV-related therapies, and the use of ZIKV as an oncolytic agent.


Asunto(s)
Apoptosis/fisiología , Infección por el Virus Zika/metabolismo , Virus Zika/fisiología , Animales , Antivirales/uso terapéutico , Muerte Celular/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Interferones/uso terapéutico , Microcefalia/virología , Fenómenos Fisiológicos de los Virus/inmunología , Replicación Viral/fisiología , Virus Zika/genética , Virus Zika/patogenicidad , Infección por el Virus Zika/virología
3.
PLoS Biol ; 19(9): e3001352, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491982

RESUMEN

Antiviral defenses can sense viral RNAs and mediate their destruction. This presents a challenge for host cells since they must destroy viral RNAs while sparing the host mRNAs that encode antiviral effectors. Here, we show that highly upregulated interferon-stimulated genes (ISGs), which encode antiviral proteins, have distinctive nucleotide compositions. We propose that self-targeting by antiviral effectors has selected for ISG transcripts that occupy a less self-targeted sequence space. Following interferon (IFN) stimulation, the CpG-targeting antiviral effector zinc-finger antiviral protein (ZAP) reduces the mRNA abundance of multiple host transcripts, providing a mechanistic explanation for the repression of many (but not all) interferon-repressed genes (IRGs). Notably, IRGs tend to be relatively CpG rich. In contrast, highly upregulated ISGs tend to be strongly CpG suppressed. Thus, ZAP is an example of an effector that has not only selected compositional biases in viral genomes but also appears to have notably shaped the composition of host transcripts in the vertebrate interferome.


Asunto(s)
Fosfatos de Dinucleósidos , Factores Reguladores del Interferón/genética , ARN Viral , Proteínas de Unión al ARN/metabolismo , Células A549 , Línea Celular , Humanos , Interferón beta/farmacología , ARN Mensajero , Proteínas de Unión al ARN/genética , Fenómenos Fisiológicos de los Virus , Virus
4.
J Gen Virol ; 102(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34424156

RESUMEN

Viruses may exploit the cardiovascular system to facilitate transmission or within-host dissemination, and the symptoms of many viral diseases stem at least in part from a loss of vascular integrity. The microvascular architecture is comprised of an endothelial cell barrier ensheathed by perivascular cells (pericytes). Pericytes are antigen-presenting cells (APCs) and play crucial roles in angiogenesis and the maintenance of microvascular integrity through complex reciprocal contact-mediated and paracrine crosstalk with endothelial cells. We here review the emerging ways that viruses interact with pericytes and pay consideration to how these interactions influence microvascular function and viral pathogenesis. Major outcomes of virus-pericyte interactions include vascular leakage or haemorrhage, organ tropism facilitated by barrier disruption, including viral penetration of the blood-brain barrier and placenta, as well as inflammatory, neurological, cognitive and developmental sequelae. The underlying pathogenic mechanisms may include direct infection of pericytes, pericyte modulation by secreted viral gene products and/or the dysregulation of paracrine signalling from or to pericytes. Viruses we cover include the herpesvirus human cytomegalovirus (HCMV, Human betaherpesvirus 5), the retrovirus human immunodeficiency virus (HIV; causative agent of acquired immunodeficiency syndrome, AIDS, and HIV-associated neurocognitive disorder, HAND), the flaviviruses dengue virus (DENV), Japanese encephalitis virus (JEV) and Zika virus (ZIKV), and the coronavirus severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2; causative agent of coronavirus disease 2019, COVID-19). We touch on promising pericyte-focussed therapies for treating the diseases caused by these important human pathogens, many of which are emerging viruses or are causing new or long-standing global pandemics.


Asunto(s)
Fenómenos Fisiológicos Celulares , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno , Pericitos/virología , Virosis/metabolismo , Virosis/virología , Animales , Comunicación Celular , Virus del Dengue/fisiología , Manejo de la Enfermedad , Células Endoteliales/virología , Endotelio/metabolismo , Endotelio/virología , VIH/fisiología , Humanos , Comunicación Paracrina , SARS-CoV-2/fisiología , Virosis/diagnóstico , Virosis/terapia , Fenómenos Fisiológicos de los Virus
5.
Viruses ; 13(8)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34452433

RESUMEN

Cell-to-cell variability of infection has long been known, yet it has remained one of the least understood phenomena in infection research. It impacts on disease onset and development, yet only recently underlying mechanisms have been studied in clonal cell cultures by single-virion immunofluorescence microscopy and flow cytometry. In this review, we showcase how single-cell RNA sequencing (scRNA-seq), single-molecule RNA-fluorescence in situ hybridization (FISH), and copper(I)-catalyzed azide-alkyne cycloaddition (click) with alkynyl-tagged viral genomes dissect infection variability in human and mouse cells. We show how the combined use of scRNA-FISH and click-chemistry reveals highly variable onsets of adenoviral gene expression, and how single live cell plaques reveal lytic and nonlytic adenovirus transmissions. The review highlights how scRNA-seq profiling and scRNA-FISH of coxsackie, influenza, dengue, zika, and herpes simplex virus infections uncover transcriptional variability, and how the host interferon response tunes influenza and sendai virus infections. We introduce the concept of "cell state" in infection variability, and conclude with advances by single-cell simultaneous measurements of chromatin accessibility and mRNA counts at high-throughput. Such technology will further dissect the sequence of events in virus infection and pathology, and better characterize the genetic and genomic stability of viruses, cell autonomous innate immune responses, and mechanisms of tissue injury.


Asunto(s)
Análisis de la Célula Individual , Virosis/metabolismo , Fenómenos Fisiológicos de los Virus , Virus/genética , Animales , Química Clic/métodos , Genoma Viral , Humanos , Inmunidad Innata , Hibridación Fluorescente in Situ/métodos , Ratones , Virión/genética , Virión/metabolismo , Virosis/virología , Replicación Viral , Virus/clasificación , Virus/metabolismo
6.
Viruses ; 13(6)2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-34203080

RESUMEN

Viruses are pathogens that have evolved to hijack the cellular machinery to replicate themselves and spread to new cells. During the course of evolution, viruses developed different strategies to overcome the cellular defenses and create new progeny. Among them, some RNA and many DNA viruses require access to the nucleus to replicate their genome. In non-dividing cells, viruses can only access the nucleus through the nuclear pore complex (NPC). Therefore, viruses have developed strategies to usurp the nuclear transport machinery and gain access to the nucleus. The majority of these viruses use the capsid to manipulate the nuclear import machinery. However, the particular tactics employed by each virus to reach the host chromatin compartment are very different. Nevertheless, they all require some degree of capsid remodeling. Recent notions on the interplay between the viral capsid and cellular factors shine new light on the quest for the nuclear entry step and for the fate of these viruses. In this review, we describe the main components and function of nuclear transport machinery. Next, we discuss selected examples of RNA and DNA viruses (HBV, HSV, adenovirus, and HIV) that remodel their capsid as part of their strategies to access the nucleus and to replicate.


Asunto(s)
Cápside/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virología , Interacciones Microbiota-Huesped , Virus/metabolismo , Transporte Activo de Núcleo Celular , Humanos , Poro Nuclear/virología , Virión/metabolismo , Fenómenos Fisiológicos de los Virus , Replicación Viral
7.
Cells ; 10(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34200500

RESUMEN

The host nucleocytoplasmic trafficking system is often hijacked by viruses to accomplish their replication and to suppress the host immune response. Viruses encode many factors that interact with the host nuclear transport receptors (NTRs) and the nucleoporins of the nuclear pore complex (NPC) to access the host nucleus. In this review, we discuss the viral factors and the host factors involved in the nuclear import and export of viral components. As nucleocytoplasmic shuttling is vital for the replication of many viruses, we also review several drugs that target the host nuclear transport machinery and discuss their feasibility for use in antiviral treatment.


Asunto(s)
Núcleo Celular/metabolismo , Núcleo Celular/virología , SARS-CoV-2/fisiología , Fenómenos Fisiológicos de los Virus , Replicación Viral/fisiología , Transporte Activo de Núcleo Celular/fisiología , COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Internalización del Virus , Virus/patogenicidad
8.
Virology ; 561: 6-16, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34089997

RESUMEN

Based on the previously reported 13-residue antibacterial peptide analog, brevinin-1EMa (FLGWLFKVASKVL, peptide B), we attempted to design a novel class of antiviral peptides. For this goal, we synthesized three peptides with different stapling positions (B-2S, B-8S, and B-5S). The most active antiviral peptide with the specific stapling position (B-5S) was further modified in combination with either cysteine (B-5S3C, B-5S7C, and B-5S10C) or hydrophilic amino acid substitution (Bsub and Bsub-5S). Overall, B, B-5S, and Bsub-5S peptides showed superior antiviral activities against enveloped viruses such as retrovirus, lentivirus, hepatitis C virus, and herpes simplex virus with EC50 values of 1-5 µM. Murine norovirus, a non-enveloped virus, was not susceptible to the virucidal actions of these peptides, suggesting the virus membrane disruption as their main antiviral mechanisms of action. We believe that these three novel peptides could serve as promising candidates for further development of membrane-targeting antiviral drugs in the future.


Asunto(s)
Antivirales/farmacología , Canales Iónicos/química , Canales Iónicos/farmacología , Péptidos/farmacología , Internalización del Virus/efectos de los fármacos , Virus/efectos de los fármacos , Antivirales/química , Antivirales/metabolismo , Bacterias/efectos de los fármacos , Línea Celular , Diseño de Fármacos , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Humanos , Canales Iónicos/metabolismo , Lentivirus/efectos de los fármacos , Lentivirus/fisiología , Pruebas de Sensibilidad Microbiana , Norovirus/efectos de los fármacos , Norovirus/fisiología , Péptidos/química , Péptidos/metabolismo , Retroviridae/efectos de los fármacos , Retroviridae/fisiología , Fenómenos Fisiológicos de los Virus
9.
Viruses ; 13(5)2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946963

RESUMEN

Viruses are infectious agents that hijack the host cell machinery in order to replicate and generate progeny. Viral infection is initiated by attachment to host cell receptors, and typical viral receptors are cell-surface-borne molecules such as proteins or glycan structures. Sialylated glycans (glycans bearing sialic acids) and glycosaminoglycans (GAGs) represent major classes of carbohydrate receptors and have been implicated in facilitating viral entry for many viruses. As interactions between viruses and sialic acids have been extensively reviewed in the past, this review provides an overview of the current state of structural knowledge about interactions between non-enveloped human viruses and GAGs. We focus here on adeno-associated viruses, human papilloma viruses (HPVs), and polyomaviruses, as at least some structural information about the interactions of these viruses with GAGs is available. We also discuss the multivalent potential for GAG binding, highlighting the importance of charged interactions and positively charged amino acids at the binding sites, and point out challenges that remain in the field.


Asunto(s)
Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Fenómenos Fisiológicos de los Virus , Animales , Humanos , Conformación Molecular , Relación Estructura-Actividad , Internalización del Virus , Virus/clasificación , Virus/metabolismo
10.
mSphere ; 6(2)2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853871

RESUMEN

Antigen recognition by the B cell receptor (BCR) is a physiological trigger for reactivation of Epstein-Barr virus (EBV) and can be recapitulated in vitro by cross-linking of surface immunoglobulins. Previously, we identified a subset of EBV microRNAs (miRNAs) that attenuate BCR signal transduction and subsequently dampen lytic reactivation in B cells. The roles of host miRNAs in the EBV lytic cycle are not completely understood. Here, we profiled the small RNAs in reactivated Burkitt lymphoma cells and identified several miRNAs, such as miR-141, that are induced upon BCR cross-linking. Notably, EBV encodes a viral miRNA, miR-BART9, with sequence homology to miR-141. To better understand the functions of these two miRNAs, we examined their molecular targets and experimentally validated multiple candidates commonly regulated by both miRNAs. Targets included B cell transcription factors and known regulators of EBV immediate-early genes, leading us to hypothesize that these miRNAs modulate kinetics of the lytic cascade in B cells. Through functional assays, we identified roles for miR-141 and EBV miR-BART9 and one specific target, FOXO3, in progression of the lytic cycle. Our data support a model whereby EBV exploits BCR-responsive miR-141 and further mimics activity of this miRNA family via a viral miRNA to promote productive lytic replication.IMPORTANCE EBV is a human pathogen associated with several malignancies. A key aspect of lifelong virus persistence is the ability to switch between latent and lytic replication modes. The mechanisms governing latency, reactivation, and progression of the lytic cycle are only partly understood. This study reveals that specific miRNAs can act to support the EBV lytic phase following BCR-mediated reactivation triggers. Furthermore, this study identifies a role for FOXO3, commonly suppressed by both host and viral miRNAs, in modulating progression of the EBV lytic cycle.


Asunto(s)
Proteína Forkhead Box O3/antagonistas & inhibidores , Proteína Forkhead Box O3/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , MicroARNs/genética , MicroARNs/inmunología , Receptores de Antígenos de Linfocitos B/genética , Activación Viral/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Línea Celular , Proteína Forkhead Box O3/inmunología , Células HEK293 , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Activación Viral/genética , Fenómenos Fisiológicos de los Virus
11.
Transl Res ; 237: 98-123, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33905949

RESUMEN

Oncolytic virotherapy is a therapeutic approach that uses replication-competent viruses to kill cancers. The ability of oncolytic viruses to selectively replicate in cancer cells leads to direct cell lysis and induction of anticancer immune response. Like other anticancer therapies, oncolytic virotherapy has several limitations such as viral delivery to the target, penetration into the tumor mass, and antiviral immune responses. This review provides an insight into the different characteristics of oncolytic viruses (natural and genetically modified) that contribute to effective applications of oncolytic virotherapy in preclinical and clinical trials, and strategies to overcome the limitations. The potential of oncolytic virotherapy combining with other conventional treatments or cancer immunotherapies involving immune checkpoint inhibitors and CAR-T therapy could form part of future multimodality treatment strategies.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Humanos , Inmunoterapia Adoptiva , Fenómenos Fisiológicos de los Virus , Virus/clasificación
12.
Science ; 371(6536)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33766858

RESUMEN

Microbial roles in cancer formation, diagnosis, prognosis, and treatment have been disputed for centuries. Recent studies have provocatively claimed that bacteria, viruses, and/or fungi are pervasive among cancers, key actors in cancer immunotherapy, and engineerable to treat metastases. Despite these findings, the number of microbes known to directly cause carcinogenesis remains small. Critically evaluating and building frameworks for such evidence in light of modern cancer biology is an important task. In this Review, we delineate between causal and complicit roles of microbes in cancer and trace common themes of their influence through the host's immune system, herein defined as the immuno-oncology-microbiome axis. We further review evidence for intratumoral microbes and approaches that manipulate the host's gut or tumor microbiome while projecting the next phase of experimental discovery.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Microbiota , Neoplasias/microbiología , Neoplasias/terapia , Inmunidad Adaptativa , Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Bacterias/genética , Carcinogénesis , Microbioma Gastrointestinal , Ingeniería Genética , Interacciones Microbiota-Huesped , Humanos , Inmunomodulación , Inmunoterapia , Tejido Linfoide/inmunología , Neoplasias/inmunología , Viroterapia Oncolítica , Microambiente Tumoral , Fenómenos Fisiológicos de los Virus
13.
Viruses ; 13(3)2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669141

RESUMEN

Viruses are highly dependent on the host they infect. Their dependence triggers processes of virus-host co-adaptation, enabling viruses to explore host resources whilst escaping immunity. Scientists have tackled viral-host interplay at differing levels of complexity-in individual hosts, organs, tissues and cells-and seminal studies advanced our understanding about viral lifecycles, intra- or inter-species transmission, and means to control infections. Recently, it emerged as important to address the physical properties of the materials in biological systems; membrane-bound organelles are only one of many ways to separate molecules from the cellular milieu. By achieving a type of compartmentalization lacking membranes known as biomolecular condensates, biological systems developed alternative mechanisms of controlling reactions. The identification that many biological condensates display liquid properties led to the proposal that liquid-liquid phase separation (LLPS) drives their formation. The concept of LLPS is a paradigm shift in cellular structure and organization. There is an unprecedented momentum to revisit long-standing questions in virology and to explore novel antiviral strategies. In the first part of this review, we focus on the state-of-the-art about biomolecular condensates. In the second part, we capture what is known about RNA virus-phase biology and discuss future perspectives of this emerging field in virology.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Fenómenos Fisiológicos de los Virus , Animales , Fenómenos Biofísicos , VIH/fisiología , Humanos , Virus de la Influenza A/fisiología , Morbillivirus/fisiología , Orgánulos/virología , SARS-CoV-2/fisiología , Vesiculovirus/fisiología , Virosis/virología , Internalización del Virus
14.
Viruses ; 13(1)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33451128

RESUMEN

Within the family of Retroviridae, foamy viruses (FVs) are unique and unconventional with respect to many aspects in their molecular biology, including assembly and release of enveloped viral particles. Both components of the minimal assembly and release machinery, Gag and Env, display significant differences in their molecular structures and functions compared to the other retroviruses. This led to the placement of FVs into a separate subfamily, the Spumaretrovirinae. Here, we describe the molecular differences in FV Gag and Env, as well as Pol, which is translated as a separate protein and not in an orthoretroviral manner as a Gag-Pol fusion protein. This feature further complicates FV assembly since a specialized Pol encapsidation strategy via a tripartite Gag-genome-Pol complex is used. We try to relate the different features and specific interaction patterns of the FV Gag, Pol, and Env proteins in order to develop a comprehensive and dynamic picture of particle assembly and release, but also other features that are indirectly affected. Since FVs are at the root of the retrovirus tree, we aim at dissecting the unique/specialized features from those shared among the Spuma- and Orthoretrovirinae. Such analyses may shed light on the evolution and characteristics of virus envelopment since related viruses within the Ortervirales, for instance LTR retrotransposons, are characterized by different levels of envelopment, thus affecting the capacity for intercellular transmission.


Asunto(s)
Infecciones por Retroviridae/virología , Spumavirus/fisiología , Ensamble de Virus , Fenómenos Fisiológicos de los Virus , Cápside/metabolismo , Genoma Viral , Interacciones Huésped-Patógeno , Humanos , Spumavirus/ultraestructura , Proteínas Virales/metabolismo , Liberación del Virus , Replicación Viral
15.
Cell Microbiol ; 23(2): e13276, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33037857

RESUMEN

Viruses confiscate cellular components of the ubiquitin-proteasome system (UPS) to facilitate many aspects of the infectious cycle. The 26S proteasome is an ATP-dependent, multisubunit proteolytic machine present in all eukaryotic cells. The proteasome executes the controlled degradation of functional proteins, as well as the hydrolysis of aberrantly folded polypeptides. There is growing evidence for the role of the UPS in viral entry. The UPS assists in several steps of the initiation of infection, including endosomal escape of the entering virion, intracellular transport of incoming nucleocapsids and uncoating of the viral genome. Inhibitors of proteasome activity, including MG132, epoxomicin, lactacystin and bortezomib have been integral to developments in this area. Here, we review the mechanistic details of UPS involvement in the entry process of viruses from a multitude of families. The possibility of proteasome inhibitors as therapeutic antiviral agents is highlighted.


Asunto(s)
Interacciones Microbiota-Huesped , Complejo de la Endopetidasa Proteasomal/fisiología , Inhibidores de Proteasoma/farmacología , Ubiquitina/fisiología , Internalización del Virus , Fenómenos Fisiológicos de los Virus , Virus/efectos de los fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Antivirales/farmacología , Bortezomib/farmacología , Humanos , Leupeptinas/farmacología , Nucleocápside/metabolismo , Oligopéptidos/farmacología , Proteolisis , Virión/metabolismo
16.
Sensors (Basel) ; 21(1)2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33374523

RESUMEN

Cell-based assays are essential for virus functional characterization in fundamental and applied research. Overcoming the limitations of virus-labelling strategies while allowing functional assessment of critical viral enzymes, virus-induced cell-based biosensors constitute a powerful approach. Herein, we designed and characterized different cell-based switch-on split GFP sensors reporting viral proteolytic activity and virus infection. Crucial to these sensors is the effective-yet reversible-fluorescence off-state, through protein distortion. For that, single (protein embedment or intein-mediated cyclization) or dual (coiled-coils) distortion schemes prevent split GFP self-assembly, until virus-promoted proteolysis of a cleavable sequence. All strategies showed their applicability in detecting viral proteolysis, although with different efficiencies depending on the protease. While for tobacco etch virus protease the best performing sensor was based on coiled-coils (signal-to-noise ratio, SNR, 97), for adenovirus and lentivirus proteases it was based on GFP11 cyclization (SNR 3.5) or GFP11 embedment distortion (SNR 6.0), respectively. When stably expressed, the sensors allowed live cell biosensing of adenovirus infection, with sensor fluorescence activation 24 h post-infection. The structural distortions herein studied are highly valuable in the development of cellular biosensing platforms. Additionally highlighted, selection of the best performing strategy is highly dependent on the unique properties of each viral protease.


Asunto(s)
Técnicas Biosensibles , Inteínas , Proteolisis , Proteínas Fluorescentes Verdes , Fenómenos Fisiológicos de los Virus
17.
Front Immunol ; 11: 593901, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240281

RESUMEN

The skin is an active immune organ that functions as the first and largest site of defense to the outside environment. Serving as the primary interface between host and pathogen, the skin's early immune responses to viral invaders often determine the course and severity of infection. We review the current literature pertaining to the mechanisms of cutaneous viral invasion for classical skin-tropic, oncogenic, and vector-borne skin viruses. We discuss the skin's evolved mechanisms for innate immune viral defense against these invading pathogens, as well as unique strategies utilized by the viruses to escape immune detection. We additionally explore the roles that demographic and environmental factors, such as age, biological sex, and the cutaneous microbiome, play in altering the host immune response to viral threats.


Asunto(s)
Dermatitis/etiología , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Inmunidad Innata , Virosis/etiología , Dermatitis/metabolismo , Ambiente , Humanos , Factores de Riesgo , Tropismo Viral , Virosis/metabolismo , Virosis/transmisión , Fenómenos Fisiológicos de los Virus , Virus/clasificación , Virus/inmunología
18.
J Virol ; 95(2)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33115868

RESUMEN

Phosphatidylserine (PS) receptors mediate clearance of apoptotic cells-efferocytosis-by recognizing the PS exposed on those cells. They also mediate the entry of enveloped viruses by binding PS in the virion membrane. Here, we show that phosphatidylethanolamine (PE) synergizes with PS to enhance PS receptor-mediated efferocytosis and virus entry. The presence of PE on the same surface as PS dramatically enhances recognition of PS by PS-binding proteins such as GAS6, PROS, and TIM1. Liposomes containing both PE and PS bound to GAS6 and were engulfed by AXL-expressing cells much more efficiently than those containing PS alone. Further, infection of AXL-expressing cells by infectious Zika virus or Ebola, Chikungunya, or eastern equine encephalitis pseudoviruses was inhibited with greater efficiency by the liposomes containing both PS and PE compared to a mixture of liposomes separately composed of PS and PE. These data demonstrate that simultaneous recognition of PE and PS maximizes PS receptor-mediated virus entry and efferocytosis and underscore the important contribution of PE in these major biological processes.IMPORTANCE Phosphatidylserine (PS) and phosphatidylethanolamine (PE) are usually sequestered to the inner leaflet of the plasma membrane of the healthy eukaryotic cells. During apoptosis, these phospholipids move to the cell's outer leaflet where they are recognized by so-called PS receptors on surveilling phagocytes. Several pathogenic families of enveloped viruses hijack these PS receptors to gain entry into their target cells. Here, we show that efficiency of these processes is enhanced, namely, PE synergizes with PS to promote PS receptor-mediated virus infection and clearance of apoptotic cells. These findings deepen our understanding of how these fundamental biological processes are executed.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Virosis/metabolismo , Fenómenos Fisiológicos de los Virus , Membrana Celular/metabolismo , Células HEK293 , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Interacciones Huésped-Patógeno , Humanos , Liposomas/metabolismo , Fagocitosis , Proteína S/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Virosis/virología , Internalización del Virus , Virus/clasificación , Virus/metabolismo , Tirosina Quinasa del Receptor Axl
20.
Appl Environ Microbiol ; 86(24)2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33036988

RESUMEN

Influent wastewater and effluent wastewater at the Rya treatment plant in Gothenburg, Sweden, were continuously monitored for enteric viruses by quantitative PCR (qPCR) during 1 year. Viruses in effluent wastewater were also identified by next-generation sequencing (NGS) in samples collected during spring, early summer, and winter. Samples of incoming wastewater were collected every second week. Seasonal variations in viral concentrations in incoming wastewater were found for noroviruses GII, sapovirus, rotavirus, parechovirus, and astrovirus. Norovirus GI and GIV and Aichi virus were present in various amounts during most weeks throughout the year, while hepatitis A virus, enterovirus, and adenovirus were identified less frequently. Fluctuations in viral concentrations in incoming wastewater were related to the number of diagnosed patients. The viruses were also detected in treated wastewater, however, with a 3- to 6-log10 reduction in concentration. Seven different hepatitis E virus (HEV) strains were identified in the effluents. Five of these strains belonged to genotype 3 and have been isolated in Sweden from swine, wild boars, and humans and in drinking water. The other two strains were divergent and had not been identified previously. They were similar to strains infecting rats and humans. Surveillance of enteric viruses in wastewater is a tool for early detection and follow-up of gastroenteritis outbreaks in society and for the identification of new viruses that can cause infection in humans.IMPORTANCE Both influent wastewater and treated wastewater at a wastewater treatment plant (WWTP) contain a high variety of human viral pathogens with seasonal variability when followed for 1 year. The peak of the amount of 11 different viruses in the inlet wastewater preceded the peak of the number of diagnosed patients by 2 to 4 weeks. The treatment of wastewater reduced viral concentrations by 3 to 6 log10 Despite the treatment of wastewater, up to 5 log10 virus particles per liter were released from into the surrounding river. Hepatitis E virus (HEV) strains previously identified in drinking water and two new strains, similar to those infecting rats and humans, were identified in the treated wastewater released from the WWTP.


Asunto(s)
Metagenoma , Virus/aislamiento & purificación , Aguas Residuales/virología , Metagenómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Estaciones del Año , Fenómenos Fisiológicos de los Virus , Virus/clasificación , Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA