Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4644, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821943

RESUMEN

The SARS-CoV-2 viral infection transforms host cells and produces special organelles in many ways, and we focus on the replication organelles, the sites of replication of viral genomic RNA (vgRNA). To date, the precise cellular localization of key RNA molecules and replication intermediates has been elusive in electron microscopy studies. We use super-resolution fluorescence microscopy and specific labeling to reveal the nanoscopic organization of replication organelles that contain numerous vgRNA molecules along with the replication enzymes and clusters of viral double-stranded RNA (dsRNA). We show that the replication organelles are organized differently at early and late stages of infection. Surprisingly, vgRNA accumulates into distinct globular clusters in the cytoplasmic perinuclear region, which grow and accommodate more vgRNA molecules as infection time increases. The localization of endoplasmic reticulum (ER) markers and nsp3 (a component of the double-membrane vesicle, DMV) at the periphery of the vgRNA clusters suggests that replication organelles are encapsulated into DMVs, which have membranes derived from the host ER. These organelles merge into larger vesicle packets as infection advances. Precise co-imaging of the nanoscale cellular organization of vgRNA, dsRNA, and viral proteins in replication organelles of SARS-CoV-2 may inform therapeutic approaches that target viral replication and associated processes.


Asunto(s)
Retículo Endoplásmico , Orgánulos , ARN Viral , SARS-CoV-2 , Replicación Viral , SARS-CoV-2/fisiología , SARS-CoV-2/ultraestructura , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , ARN Viral/metabolismo , ARN Viral/genética , Replicación Viral/fisiología , Humanos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Retículo Endoplásmico/ultraestructura , Orgánulos/virología , Orgánulos/metabolismo , Orgánulos/ultraestructura , Chlorocebus aethiops , Células Vero , Animales , COVID-19/virología , COVID-19/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Microscopía Fluorescente , Compartimentos de Replicación Viral/metabolismo , ARN Bicatenario/metabolismo
2.
J Cell Biol ; 222(7)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37093123

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the etiologic agent for the global COVID-19 pandemic, triggers the formation of endoplasmic reticulum (ER)-derived replication organelles, including double-membrane vesicles (DMVs), in the host cell to support viral replication. Here, we clarify how SARS-CoV-2 hijacks host factors to construct the DMVs. We show that the ER morphogenic proteins reticulon-3 (RTN3) and RTN4 help drive DMV formation, enabling viral replication, which leads to productive infection. Different SARS-CoV-2 variants, including the delta variant, use the RTN-dependent pathway to promote infection. Mechanistically, our results reveal that the membrane-embedded reticulon homology domain (RHD) of the RTNs is sufficient to functionally support viral replication and physically engage NSP3 and NSP4, two viral non-structural membrane proteins known to induce DMV formation. Our findings thus identify the ER morphogenic RTN3 and RTN4 membrane proteins as host factors that help promote the biogenesis of SARS-CoV-2-induced DMVs, which can act as viral replication platforms.


Asunto(s)
Retículo Endoplásmico , Proteínas de la Membrana , Orgánulos , SARS-CoV-2 , Humanos , COVID-19/virología , Retículo Endoplásmico/virología , Proteínas de la Membrana/metabolismo , Pandemias , SARS-CoV-2/fisiología , Replicación Viral , Orgánulos/virología , Proteínas no Estructurales Virales/metabolismo
3.
J Mol Biol ; 435(16): 167988, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-36709795

RESUMEN

The past decade has seen a revolution in our understanding of how the cellular environment is organized, where an incredible body of work has provided new insights into the role played by membraneless organelles. These rapid advancements have been made possible by an increasing awareness of the peculiar physical properties that give rise to such bodies and the complex biology that enables their function. Viral infections are not extraneous to this. Indeed, in host cells, viruses can harness existing membraneless compartments or, even, induce the formation of new ones. By hijacking the cellular machinery, these intracellular bodies can assist in the replication, assembly, and packaging of the viral genome as well as in the escape of the cellular immune response. Here, we provide a perspective on the fundamental polymer physics concepts that may help connect and interpret the different observed phenomena, ranging from the condensation of viral genomes to the phase separation of multicomponent solutions. We complement the discussion of the physical basis with a description of biophysical methods that can provide quantitative insights for testing and developing theoretical and computational models.


Asunto(s)
Condensados Biomoleculares , Biopolímeros , Orgánulos , Empaquetamiento del Genoma Viral , Virosis , Replicación Viral , Humanos , Orgánulos/química , Orgánulos/virología , Virosis/virología , Condensados Biomoleculares/virología , Gránulos de Estrés/química , Gránulos de Estrés/virología , Genoma Viral , Biopolímeros/química , Transición de Fase
4.
PLoS Pathog ; 17(12): e1010113, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34871328

RESUMEN

Emerging coronaviruses (CoVs) pose a severe threat to human and animal health worldwide. To identify host factors required for CoV infection, we used α-CoV transmissible gastroenteritis virus (TGEV) as a model for genome-scale CRISPR knockout (KO) screening. Transmembrane protein 41B (TMEM41B) was found to be a bona fide host factor involved in infection by CoV and three additional virus families. We found that TMEM41B is critical for the internalization and early-stage replication of TGEV. Notably, our results also showed that cells lacking TMEM41B are unable to form the double-membrane vesicles necessary for TGEV replication, indicating that TMEM41B contributes to the formation of CoV replication organelles. Lastly, our data from a mouse infection model showed that the KO of this factor can strongly inhibit viral infection and delay the progression of a CoV disease. Our study revealed that targeting TMEM41B is a highly promising approach for the development of broad-spectrum anti-viral therapeutics.


Asunto(s)
Sistemas CRISPR-Cas , Gastroenteritis Porcina Transmisible/virología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/fisiología , Orgánulos/virología , Virus de la Gastroenteritis Transmisible/fisiología , Replicación Viral , Animales , Gastroenteritis Porcina Transmisible/genética , Gastroenteritis Porcina Transmisible/transmisión , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Porcinos
5.
Viruses ; 13(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34578359

RESUMEN

A common viral replication strategy is characterized by the assembly of intracellular compartments that concentrate factors needed for viral replication and simultaneously conceal the viral genome from host-defense mechanisms. Recently, various membrane-less virus-induced compartments and cellular organelles have been shown to represent biomolecular condensates (BMCs) that assemble through liquid-liquid phase separation (LLPS). In the present work, we analyze biophysical properties of intranuclear replication compartments (RCs) induced during human adenovirus (HAdV) infection. The viral ssDNA-binding protein (DBP) is a major component of RCs that contains intrinsically disordered and low complexity proline-rich regions, features shared with proteins that drive phase transitions. Using fluorescence recovery after photobleaching (FRAP) and time-lapse studies in living HAdV-infected cells, we show that DBP-positive RCs display properties of liquid BMCs, which can fuse and divide, and eventually form an intranuclear mesh with less fluid-like features. Moreover, the transient expression of DBP recapitulates the assembly and liquid-like properties of RCs in HAdV-infected cells. These results are of relevance as they indicate that DBP may be a scaffold protein for the assembly of HAdV-RCs and should contribute to future studies on the role of BMCs in virus-host cell interactions.


Asunto(s)
Adenoviridae/metabolismo , Condensados Biomoleculares , Proteínas de Unión al ADN/metabolismo , Compartimentos de Replicación Viral/fisiología , Replicación Viral/fisiología , Adenoviridae/genética , Infecciones por Adenoviridae , Adenovirus Humanos/metabolismo , Línea Celular , Proteínas de Unión al ADN/química , Interacciones Microbiota-Huesped , Humanos , Orgánulos/virología , Dominios Proteicos , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
6.
Cells ; 10(9)2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34572055

RESUMEN

All intracellular pathogens critically depend on host cell organelles and metabolites for successful infection and replication. One hallmark of positive-strand RNA viruses is to induce alterations of the (endo)membrane system in order to shield their double-stranded RNA replication intermediates from detection by the host cell's surveillance systems. This spatial seclusion also allows for accruing host and viral factors and building blocks required for efficient replication of the genome and prevents access of antiviral effectors. Even though the principle is iterated by almost all positive-strand RNA viruses infecting plants and animals, the specific structure and the organellar source of membranes differs. Here, we discuss the characteristic ultrastructural features of the virus-induced membranous replication organelles in plant and animal cells and the scientific progress gained by advanced microscopy methods.


Asunto(s)
Interacciones Huésped-Patógeno , Membranas Intracelulares/ultraestructura , Orgánulos/ultraestructura , Virus ARN Monocatenarios Positivos/patogenicidad , Infecciones por Virus ARN/patología , ARN Viral/genética , Replicación Viral , Animales , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virología , Orgánulos/metabolismo , Orgánulos/virología , Plantas , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología
7.
J Virol ; 95(21): e0107621, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34406861

RESUMEN

Positive-strand RNA viruses induce the biogenesis of unique membranous organelles called viral replication organelles (VROs), which perform virus replication in infected cells. Tombusviruses have been shown to rewire cellular trafficking and metabolic pathways, remodel host membranes, and recruit multiple host factors to support viral replication. In this work, we demonstrate that tomato bushy stunt virus (TBSV) and the closely related carnation Italian ringspot virus (CIRV) usurp Rab7 small GTPase to facilitate building VROs in the surrogate host yeast and in plants. Depletion of Rab7 small GTPase, which is needed for late endosome and retromer biogenesis, strongly inhibits TBSV and CIRV replication in yeast and in planta. The viral p33 replication protein interacts with Rab7 small GTPase, which results in the relocalization of Rab7 into the large VROs. Similar to the depletion of Rab7, the deletion of either MON1 or CCZ1 heterodimeric GEFs (guanine nucleotide exchange factors) of Rab7 inhibited TBSV RNA replication in yeast. This suggests that the activated Rab7 has proviral functions. We show that the proviral function of Rab7 is to facilitate the recruitment of the retromer complex and the endosomal sorting nexin-BAR proteins into VROs. We demonstrate that TBSV p33-driven retargeting of Rab7 into VROs results in the delivery of several retromer cargos with proviral functions. These proteins include lipid enzymes, such as Vps34 PI3K (phosphatidylinositol 3-kinase), PI4Kα-like Stt4 phosphatidylinositol 4-kinase, and Psd2 phosphatidylserine decarboxylase. In summary, based on these and previous findings, we propose that subversion of Rab7 into VROs allows tombusviruses to reroute endocytic and recycling trafficking to support virus replication. IMPORTANCE The replication of positive-strand RNA viruses depends on the biogenesis of viral replication organelles (VROs). However, the formation of membranous VROs is not well understood yet. Using tombusviruses and the model host yeast, we discovered that the endosomal Rab7 small GTPase is critical for the formation of VROs. Interaction between Rab7 and the TBSV p33 replication protein leads to the recruitment of Rab7 into VROs. TBSV-driven usurping of Rab7 has proviral functions through facilitating the delivery of the co-opted retromer complex, sorting nexin-BAR proteins, and lipid enzymes into VROs to create an optimal milieu for virus replication. These results open up the possibility that controlling cellular Rab7 activities in infected cells could be a target for new antiviral strategies.


Asunto(s)
Nicotiana/virología , Orgánulos/virología , Saccharomyces cerevisiae/virología , Tombusvirus/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Proteínas de Unión al GTP rab/fisiología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Endosomas/metabolismo , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/fisiología , Interacciones Microbiota-Huesped , Orgánulos/metabolismo , Enfermedades de las Plantas/virología , Unión Proteica , Transporte de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Nexinas de Clasificación/metabolismo
8.
J Virol ; 95(21): e0131021, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379504

RESUMEN

Dengue virus (DENV) constitutes one of the most important arboviral pathogens affecting humans. The high prevalence of DENV infections, which cause more than 20,000 deaths annually, and the lack of effective vaccines or direct-acting antiviral drugs make it a global health concern. DENV genome replication occurs in close association with the host endomembrane system, which is remodeled to form the viral replication organelle that originates from endoplasmic reticulum (ER) membranes. To date, the viral and cellular determinants responsible for the biogenesis of DENV replication organelles are still poorly defined. The viral nonstructural protein 4A (NS4A) can remodel membranes and has been shown to associate with numerous host factors in DENV-replicating cells. In the present study, we used reverse and forward genetic screens and identified sites within NS4A required for DENV replication. We also mapped the determinants in NS4A required for interactions with other viral proteins. Moreover, taking advantage of our recently developed polyprotein expression system, we evaluated the role of NS4A in the formation of DENV replication organelles. Together, we report a detailed map of determinants within NS4A required for RNA replication, interaction with other viral proteins, and replication organelle formation. Our results suggest that NS4A might be an attractive target for antiviral therapy. IMPORTANCE DENV is the most prevalent mosquito-borne virus, causing around 390 million infections each year. There are no approved therapies to treat DENV infection, and the only available vaccine shows limited efficacy. The viral nonstructural proteins have emerged as attractive drug targets due to their pivotal role in RNA replication and establishment of virus-induced membranous compartments, designated replication organelles (ROs). The transmembrane protein NS4A, generated by cleavage of the NS4A-2K-4B precursor, contributes to DENV replication by unknown mechanisms. Here, we report a detailed genetic interaction map of NS4A and identify residues required for RNA replication and interaction between NS4A-2K-4B and NS2B-3 as well as NS1. Importantly, by means of an expression-based system, we demonstrate the essential role of NS4A in RO biogenesis and identify determinants in NS4A required for this process. Our data suggest that NS4A is an attractive target for antiviral therapy.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Biogénesis de Organelos , Orgánulos/virología , Proteínas no Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Virus del Dengue/ultraestructura , Interacciones Microbiota-Huesped , Humanos , Proteínas Mutantes/fisiología , Mutación , Orgánulos/ultraestructura , Unión Proteica , ARN/metabolismo , ARN Viral , Genética Inversa/métodos , Células Vero , Replicación Viral
9.
Viruses ; 13(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34452345

RESUMEN

Viral proteins interact with different sets of host cell components throughout the viral life cycle and are known to localize to the intracellular membraneless organelles (MLOs) of the host cell, where formation/dissolution is regulated by phase separation of intrinsically disordered proteins and regions (IDPs/IDRs). Viral proteins are rich in IDRs, implying that viruses utilize IDRs to regulate phase separation of the host cell organelles and augment replication by commandeering the functions of the organelles and/or sneaking into the organelles to evade the host immune response. This review aims to integrate current knowledge of the structural properties and intracellular localizations of viral IDPs to understand viral strategies in the host cell. First, the properties of viral IDRs are reviewed and similarities and differences with those of eukaryotes are described. The higher IDR content in viruses with smaller genomes suggests that IDRs are essential characteristics of viral proteins. Then, the interactions of the IDRs of flaviviruses with the MLOs of the host cell are investigated with emphasis on the viral proteins localized in the nucleoli and stress granules. Finally, the possible roles of viral IDRs in regulation of the phase separation of organelles and future possibilities for antiviral drug development are discussed.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/fisiología , Interacciones Huésped-Patógeno , Animales , Flavivirus/genética , Infecciones por Flavivirus/fisiopatología , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Orgánulos/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
mBio ; 12(4): e0140821, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34225484

RESUMEN

The function of the mammalian orthoreovirus (reovirus) σNS nonstructural protein is enigmatic. σNS is an RNA-binding protein that forms oligomers and enhances the stability of bound RNAs, but the mechanisms by which it contributes to reovirus replication are unknown. To determine the function of σNS-RNA binding in reovirus replication, we engineered σNS mutants deficient in RNA-binding capacity. We found that alanine substitutions of positively charged residues in a predicted RNA-binding domain decrease RNA-dependent oligomerization. To define steps in reovirus replication facilitated by the RNA-binding property of σNS, we established a complementation system in which wild-type or mutant forms of σNS could be tested for the capacity to overcome inhibition of σNS expression. Mutations in σNS that disrupt RNA binding also diminish viral replication and σNS distribution to viral factories. Moreover, viral mRNAs only incorporate into viral factories or factory-like structures (formed following expression of nonstructural protein µNS) when σNS is present and capable of binding RNA. Collectively, these findings indicate that σNS requires positively charged residues in a putative RNA-binding domain to recruit viral mRNAs to sites of viral replication and establish a function for σNS in reovirus replication. IMPORTANCE Viral replication requires the formation of neoorganelles in infected cells to concentrate essential viral and host components. However, for many viruses, it is unclear how these components coalesce into neoorganelles to form factories for viral replication. We discovered that two mammalian reovirus nonstructural proteins act in concert to form functioning viral factories. Reovirus µNS proteins assemble into exclusive factory scaffolds that require reovirus σNS proteins for efficient viral mRNA incorporation. Our results demonstrate a role for σNS in RNA recruitment to reovirus factories and, more broadly, show how a cytoplasmic non-membrane-enclosed factory is formed by an RNA virus. Understanding the mechanisms of viral factory formation will help identify new targets for antiviral therapeutics that disrupt assembly of these structures and inform the use of nonpathogenic viruses for biotechnological applications.


Asunto(s)
Orgánulos/virología , ARN Viral/genética , Reoviridae/genética , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Células HEK293 , Humanos , Mutación , Proteínas de Unión al ARN/genética , Reoviridae/química , Reoviridae/fisiología , Proteínas no Estructurales Virales/metabolismo
11.
Curr Opin Virol ; 48: 30-41, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33845410

RESUMEN

Positive-strand RNA viruses depend on intensive manipulation of subcellular organelles and membranes to create unique viral replication organelles (VROs), which represent the sites of robust virus replication. The host endomembrane-based protein-trafficking and vesicle-trafficking pathways are specifically targeted by many (+)RNA viruses to take advantage of their rich resources. We summarize the critical roles of co-opted endoplasmic reticulum subdomains and associated host proteins and COPII vesicles play in tombusvirus replication. We also present the surprising contribution of the early endosome and the retromer tubular transport carriers to VRO biogenesis. The central player is tomato bushy stunt virus (TBSV), which provides an outstanding system based on the identification of a complex network of interactions with the host cells. We present the emerging theme on how TBSV uses tethering and membrane-shaping proteins and lipid modifying enzymes to build the sophisticated VRO membranes with unique lipid composition.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Orgánulos/virología , Tombusvirus/fisiología , Replicación Viral/fisiología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Genes Virales/genética , Interacciones Huésped-Patógeno/genética , Metabolismo de los Lípidos , Lípidos , Magnoliopsida/virología , Virus ARN , Tombusvirus/genética , Replicación Viral/genética
12.
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
13.
Cell Microbiol ; 23(8): e13328, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33740320

RESUMEN

Annulate lamellae (AL) have been observed many times over the years on electron micrographs of rapidly dividing cells, but little is known about these unusual organelles consisting of stacked sheets of endoplasmic reticulum-derived membranes with nuclear pore complexes (NPCs). Evidence is growing for a role of AL in viral infection. AL have been observed early in the life cycles of the hepatitis C virus (HCV) and, more recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), suggesting a specific induction of mechanisms potentially useful to these pathogens. Like other positive-strand RNA viruses, these viruses induce host cells membranes rearrangements. The NPCs of AL could potentially mediate exchanges between these partially sealed compartments and the cytoplasm. AL may also be involved in regulating Ca2+ homeostasis or cell cycle control. They were recently observed in cells infected with Theileria annulata, an intracellular protozoan parasite inducing cell proliferation. Further studies are required to clarify their role in intracellular pathogen/host-cell interactions.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Orgánulos/microbiología , Orgánulos/parasitología , Animales , COVID-19 , Citoplasma/virología , Retículo Endoplásmico/microbiología , Retículo Endoplásmico/parasitología , Retículo Endoplásmico/ultraestructura , Retículo Endoplásmico/virología , Humanos , Orgánulos/ultraestructura , Orgánulos/virología , SARS-CoV-2/fisiología
14.
Cells ; 11(1)2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-35011607

RESUMEN

The lamellar body (LB) of the alveolar type II (ATII) cell is a lysosome-related organelle (LRO) that contains surfactant, a complex mix of mainly lipids and specific surfactant proteins. The major function of surfactant in the lung is the reduction of surface tension and stabilization of alveoli during respiration. Its lack or deficiency may cause various forms of respiratory distress syndrome (RDS). Surfactant is also part of the innate immune system in the lung, defending the organism against air-borne pathogens. The limiting (organelle) membrane that encloses the LB contains various transporters that are in part responsible for translocating lipids and other organic material into the LB. On the other hand, this membrane contains ion transporters and channels that maintain a specific internal ion composition including the acidic pH of about 5. Furthermore, P2X4 receptors, ligand gated ion channels of the danger signal ATP, are expressed in the limiting LB membrane. They play a role in boosting surfactant secretion and fluid clearance. In this review, we discuss the functions of these transporting pathways of the LB, including possible roles in disease and as therapeutic targets, including viral infections such as SARS-CoV-2.


Asunto(s)
COVID-19/metabolismo , Canales Iónicos/metabolismo , Cuerpos Lamelares/metabolismo , Pulmón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Surfactantes Pulmonares/metabolismo , COVID-19/virología , Humanos , Pulmón/virología , Orgánulos/metabolismo , Orgánulos/virología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/virología , SARS-CoV-2/fisiología
15.
Methods Mol Biol ; 2233: 93-100, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33222129

RESUMEN

Endocytosis is a vesicle-based mechanism by which eukaryotic cells internalize extracellular material. There are several types of this universal mechanism linked to different types of endocytosed cargo, including pathogens; therefore, several approaches can be applied. Here, we describe techniques that are applicable to study the internalization of flaviviruses; dextrans; transporters, such as, glutamate transporter vGlut1; and peptidergic signaling molecules, including atrial natriuretic peptide into astrocytes, the most heterogeneous neuroglial cells, which play a key homeostatic role in the central nervous system.


Asunto(s)
Factor Natriurético Atrial/genética , Endocitosis/genética , Biología Molecular/métodos , Transporte de Proteínas/genética , Astrocitos/metabolismo , Astrocitos/microbiología , Astrocitos/virología , Factor Natriurético Atrial/farmacología , Calcio/metabolismo , Flavivirus/efectos de los fármacos , Humanos , Orgánulos/genética , Orgánulos/metabolismo , Orgánulos/virología , Internalización del Virus/efectos de los fármacos
16.
J Hepatol ; 73(3): 549-558, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32294532

RESUMEN

BACKGROUND & AIMS: HCV is a positive-strand RNA virus that primarily infects human hepatocytes. Recent studies have reported that C19orf66 is expressed as an interferon (IFN)-stimulated gene; however, the intrinsic regulation of this gene within the liver as well as its antiviral effects against HCV remain elusive. METHODS: Expression of C19orf66 was quantified in both liver biopsies and primary human hepatocytes, with or without HCV infection. Mechanistic studies of the potent anti-HCV phenotype mediated by C19orf66 were conducted using state-of-the-art virological, biochemical and genetic approaches, as well as correlative light and electron microscopy and transcriptome and proteome analysis. RESULTS: Upregulation of C19orf66 mRNA was observed in both primary human hepatocytes upon HCV infection and in the livers of patients with chronic hepatitis C (CHC). In addition, pegIFNα/ribavirin therapy induced C19orf66 expression in patients with CHC. Transcriptomic profiling and whole cell proteomics of hepatoma cells ectopically expressing C19orf66 revealed no induction of other antiviral genes. Expression of C19orf66 restricted HCV infection, whereas CRIPSPR/Cas9 mediated knockout of C19orf66 attenuated IFN-mediated suppression of HCV replication. Co-immunoprecipitation followed by mass spectrometry identified a stress granule protein-dominated interactome of C19orf66. Studies with subgenomic HCV replicons and an expression system revealed that C19orf66 expression impairs HCV-induced elevation of phosphatidylinositol-4-phosphate, alters the morphology of the viral replication organelle (termed the membranous web) and thereby targets viral RNA replication. CONCLUSION: C19orf66 is an IFN-stimulated gene, which is upregulated in hepatocytes within the first hours post IFN treatment or HCV infection in vivo. The encoded protein possesses specific antiviral activity against HCV and targets the formation of the membranous web. Our study identifies C19orf66 as an IFN-inducible restriction factor with a novel antiviral mechanism that specifically targets HCV replication. LAY SUMMARY: Interferon-stimulated genes are thought to be important to for antiviral immune responses to HCV. Herein, we analysed C19orf66, an interferon-stimulated gene, which appears to inhibit HCV replication. It prevents the HCV-induced elevation of phosphatidylinositol-4-phosphate and alters the morphology of HCV's replication organelle.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/genética , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/metabolismo , Interferones/uso terapéutico , Orgánulos/virología , Proteínas de Unión al ARN/metabolismo , Compartimentos de Replicación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Adulto , Línea Celular Tumoral , Femenino , Técnicas de Inactivación de Genes , Genotipo , Células HEK293 , Hepatitis C Crónica/patología , Hepatitis C Crónica/virología , Hepatocitos/metabolismo , Humanos , Hígado/patología , Masculino , Persona de Mediana Edad , Orgánulos/efectos de los fármacos , Orgánulos/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Replicón/efectos de los fármacos , Replicón/genética , Ribavirina/uso terapéutico , Resultado del Tratamiento , Replicación Viral/genética
17.
Anal Biochem ; 597: 113691, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32194074

RESUMEN

Membraneless organelles (MLOs) in the cytoplasm and nucleus in the form of 2D and 3D phase-separated biomolecular condensates are increasingly viewed as critical in regulating diverse cellular functions. These functions include cell signaling, immune synapse function, nuclear transcription, RNA splicing and processing, mRNA storage and translation, virus replication and maturation, antiviral mechanisms, DNA sensing, synaptic transmission, protein turnover and mitosis. Components comprising MLOs often associate with low affinity; thus cell integrity can be critical to the maintenance of the full complement of respective MLO components. Phase-separated condensates are typically metastable (shape-changing) and can undergo dramatic, rapid and reversible assembly and disassembly in response to cell signaling events, cell stress, during mitosis, and after changes in cytoplasmic "crowding" (as observed with condensates of the human myxovirus resistance protein MxA). Increasing evidence suggests that neuron-specific aberrations in phase-separation properties of RNA-binding proteins (e.g. FUS and TDP-43) and others (such as the microtubule-binding protein tau) contribute to the development of degenerative neurological diseases (e.g. amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and Alzheimer's disease). Thus, studies of liquid-like phase separation (LLPS) and the formation, structure and function of MLOs are of considerable importance in understanding basic cell biology and the pathogenesis of human diseases.


Asunto(s)
Núcleo Celular/química , Citoplasma/química , Proteínas de Resistencia a Mixovirus/aislamiento & purificación , Orgánulos/química , Biología Celular , Núcleo Celular/virología , Citoplasma/virología , Humanos , Proteínas de Resistencia a Mixovirus/química , Orgánulos/virología
18.
Artículo en Inglés | MEDLINE | ID: mdl-31681621

RESUMEN

Regulation of RNA homeostasis or "RNAstasis" is a central step in eukaryotic gene expression. From transcription to decay, cellular messenger RNAs (mRNAs) associate with specific proteins in order to regulate their entire cycle, including mRNA localization, translation and degradation, among others. The best characterized of such RNA-protein complexes, today named membraneless organelles, are Stress Granules (SGs) and Processing Bodies (PBs) which are involved in RNA storage and RNA decay/storage, respectively. Given that SGs and PBs are generally associated with repression of gene expression, viruses have evolved different mechanisms to counteract their assembly or to use them in their favor to successfully replicate within the host environment. In this review we summarize the current knowledge about the viral regulation of SGs and PBs, which could be a potential novel target for the development of broad-spectrum antiviral therapies.


Asunto(s)
Interacciones Huésped-Patógeno , Orgánulos , Virosis/metabolismo , Virosis/virología , Fenómenos Fisiológicos de los Virus , Animales , Gránulos Citoplasmáticos , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Orgánulos/metabolismo , Orgánulos/virología , Transducción de Señal , Estrés Fisiológico , Virosis/genética , Fenómenos Fisiológicos de los Virus/efectos de los fármacos , Replicación Viral , Virus/clasificación , Virus/efectos de los fármacos , Virus/genética
19.
Viruses ; 11(11)2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31694296

RESUMEN

Porcine deltacoronavirus (PDCoV) was first identified in Hong Kong in 2012 from samples taken from pigs in 2009. PDCoV was subsequently identified in the USA in 2014 in pigs with a history of severe diarrhea. The virus has now been detected in pigs in several countries around the world. Following the development of tissue culture adapted strains of PDCoV, it is now possible to address questions regarding virus-host cell interactions for this genera of coronavirus. Here, we presented a detailed study of PDCoV-induced replication organelles. All positive-strand RNA viruses induce the rearrangement of cellular membranes during virus replication to support viral RNA synthesis, forming the replication organelle. Replication organelles for the Alpha-, Beta-, and Gammacoronavirus genera have been characterized. All coronavirus genera induced the formation of double-membrane vesicles (DMVs). In addition, Alpha- and Betacoronaviruses induce the formation of convoluted membranes, while Gammacoronaviruses induce the formation of zippered endoplasmic reticulum (ER) with tethered double-membrane spherules. However, the structures induced by Deltacoronaviruses, particularly the presence of convoluted membranes or double-membrane spherules, are unknown. Initially, the dynamics of PDCoV strain OH-FD22 replication were assessed with the onset of viral RNA synthesis, protein synthesis, and progeny particle release determined. Subsequently, virus-induced membrane rearrangements were identified in infected cells by electron microscopy. As has been observed for all other coronaviruses studied to date, PDCoV replication was found to induce the formation of double-membrane vesicles. Significantly, however, PDCoV replication was also found to induce the formation of regions of zippered endoplasmic reticulum, small associated tethered vesicles, and double-membrane spherules. These structures strongly resemble the replication organelle induced by avian Gammacoronavirus infectious bronchitis virus.


Asunto(s)
Coronavirus , Retículo Endoplásmico/ultraestructura , Membranas Intracelulares/ultraestructura , Orgánulos/ultraestructura , Replicación Viral , Animales , Línea Celular , Coronavirus/fisiología , Coronavirus/ultraestructura , Infecciones por Coronavirus/virología , Retículo Endoplásmico/virología , Interacciones Huésped-Patógeno , Membranas Intracelulares/virología , Cinética , Orgánulos/virología , ARN Viral/biosíntesis , Porcinos
20.
Adv Virus Res ; 105: 1-33, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31522702

RESUMEN

Transmission electron microscopy (TEM) has been crucial to study viral infections. As a result of recent advances in light and electron microscopy, we are starting to be aware of the variety of structures that viruses assemble inside cells. Viruses often remodel cellular compartments to build their replication factories. Remarkably, viruses are also able to induce new membranes and new organelles. Here we revise the most relevant imaging technologies to study the biogenesis of viral replication organelles. Live cell microscopy, correlative light and electron microscopy, cryo-TEM, and three-dimensional imaging methods are unveiling how viruses manipulate cell organization. In particular, methods for molecular mapping in situ in two and three dimensions are revealing how macromolecular complexes build functional replication complexes inside infected cells. The combination of all these imaging approaches is uncovering the viral life cycle events with a detail never seen before.


Asunto(s)
Interacciones Microbiota-Huesped , Microscopía Electrónica/métodos , Orgánulos/ultraestructura , Orgánulos/virología , Replicación Viral , Virus/crecimiento & desarrollo , Virus/ultraestructura , Procesamiento de Imagen Asistido por Computador , Microscopía/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA